201
|
Côté J, LeBlanc R, Mian H, Chu MP, McCurdy A, Masih-Khan E, Su J, Jimenez-Zepeda VH, Song K, Louzada M, White D, Sebag M, Reiman A, Stakiw J, Kotb R, Bergstrom D, Aslam M, Kaedbey R, Venner CP, Gul E, Reece D. Real-world results of autologous stem cell transplantation in newly diagnosed multiple myeloma: a report from the Canadian Myeloma Research Group database. Blood Cancer J 2023; 13:137. [PMID: 37669949 PMCID: PMC10480201 DOI: 10.1038/s41408-023-00905-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/07/2023] Open
Abstract
Autologous stem cell transplant (ASCT) remains an important option for eligible multiple myeloma (MM) patients as part of initial therapy. Using the Canadian Myeloma Research Group (CMRG) national database, we examined the details and outcomes of ASCT performed as first-line therapy in eligible Canadian MM patients between 2007 to 2021. We included 3821 patients with 72% receiving CyBorD induction and 2061 patients receiving maintenance, consisting of lenalidomide +/- steroids in 78.3%. The median PFS and OS for patients given a single ASCT were 35.4 and 126 months. Those receiving a second induction regimen had significantly inferior outcomes, although when maintenance was used, results were comparable regardless of the number of induction regimens administered (median PFS 55.3 vs 51.1 months [p = 0.11]; median OS 158.6 vs not yet reached [p = 0.13]). Consolidation patients had a longer median PFS (55.3 vs 34.4 months [p = 0.001]), but no significant gain in median OS (p = 0.065). Patients who received lenalidomide-based maintenance experienced a median PFS of 53.7 months and OS of 159 months. In the multivariable analysis, use of any type of maintenance therapy vs no maintenance was associated with a lower risk of progression (HR 0.52 (95% CI 0.47-0.57)) and death (HR 0.58 (95% CI 0.51-0.67)). This real-world study demonstrates that, overall, first-line treatment sequence in transplant-eligible patients produces a median OS of ≥10 years. It also highlights the contribution of post-ASCT maintenance, particularly lenalidomide given until progression.
Collapse
Affiliation(s)
- Julie Côté
- CHU de Québec-Université Laval, Quebec, QC, Canada.
| | | | - Hira Mian
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Michael P Chu
- Department of Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Arleigh McCurdy
- Department of Medicine, Division of Hematology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Esther Masih-Khan
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Jiandong Su
- Canadian Myeloma Research Group, Toronto, Ontario, Canada
| | | | - Kevin Song
- Division of Hematology, University of British Columbia and Leukemia/BMT Program of BC, Vancouver General Hospital, Vancouver, BC, Canada
| | - Martha Louzada
- University of Western Ontario, London Health Sciences Centre, London, ON, Canada
| | - Darrell White
- Division of Hematology, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | - Michael Sebag
- Division of Hematology, McGill University Health Centre, Montreal, QC, Canada
| | | | | | - Rami Kotb
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Debra Bergstrom
- Division of Hematology, Memorial University of Newfoundland, Newfoundland and Labrador, St John's, NL, Canada
| | | | - Rayan Kaedbey
- Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Christopher P Venner
- Lymphoma and Myeloma Program, BC Cancer, Vancouver Centre, Vancouver, BC, Canada
| | - Engin Gul
- Canadian Myeloma Research Group, Toronto, ON, Canada
| | - Donna Reece
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
202
|
Coffey DG, Maura F, Gonzalez-Kozlova E, Diaz-Mejia JJ, Luo P, Zhang Y, Xu Y, Warren EH, Dawson T, Lee B, Xie H, Smith E, Ciardiello A, Cho HJ, Rahman A, Kim-Schulze S, Diamond B, Lesokhin A, Kazandjian D, Pugh TJ, Green DJ, Gnjatic S, Landgren O. Immunophenotypic correlates of sustained MRD negativity in patients with multiple myeloma. Nat Commun 2023; 14:5335. [PMID: 37660077 PMCID: PMC10475030 DOI: 10.1038/s41467-023-40966-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/18/2023] [Indexed: 09/04/2023] Open
Abstract
The role of the immune microenvironment in maintaining disease remission in patients with multiple myeloma (MM) is not well understood. In this study, we comprehensively profile the immune system in patients with newly diagnosed MM receiving continuous lenalidomide maintenance therapy with the aim of discovering correlates of long-term treatment response. Leveraging single-cell RNA sequencing and T cell receptor β sequencing of the peripheral blood and CyTOF mass cytometry of the bone marrow, we longitudinally characterize the immune landscape in 23 patients before and one year after lenalidomide exposure. We compare patients achieving sustained minimal residual disease (MRD) negativity to patients who never achieved or were unable to maintain MRD negativity. We observe that the composition of the immune microenvironment in both the blood and the marrow varied substantially according to both MRD negative status and history of autologous stem cell transplant, supporting the hypothesis that the immune microenvironment influences the depth and duration of treatment response.
Collapse
Affiliation(s)
- David G Coffey
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Francesco Maura
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - J Javier Diaz-Mejia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ping Luo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yong Zhang
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Yuexin Xu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Edus H Warren
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Travis Dawson
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Lee
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hui Xie
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric Smith
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amanda Ciardiello
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hearn J Cho
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Multiple Myeloma Research Foundation, Norwalk, USA
| | - Adeeb Rahman
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Benjamin Diamond
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Alexander Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dickran Kazandjian
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ola Landgren
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
203
|
Liu J, Yan W, Fan H, Xu J, Li L, Du C, Mao X, Yan Y, Xu Y, Sui W, Deng S, Yi S, Anderson KC, Qiu L, Zou D, An G. Clinical Benefit of Autologous Stem Cell Transplantation for Patients with Multiple Myeloma Achieving Undetectable Minimal Residual Disease after Induction Treatment. CANCER RESEARCH COMMUNICATIONS 2023; 3:1770-1780. [PMID: 37680953 PMCID: PMC10481879 DOI: 10.1158/2767-9764.crc-23-0185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/10/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023]
Abstract
Attaining undetectable minimal residual disease (MRD) is the current therapeutic goal for multiple myeloma. But there is a current lack of data regarding the clinical benefit of autologous stem cell transplantation (ASCT) for patients with myeloma achieving early MRD-negative status after induction treatment, in addition to the interaction of longitudinal MRD status with ASCT. The current study included 407 patients with transplant-eligible multiple myeloma with available MRD status from the National Longitudinal Cohort of Hematological Diseases in China (NCT04645199), of whom 147 (34.4%) achieved early undetectable MRD and 182 (44.7%) received ASCT. Early MRD-negative status was associated with a lower risk of disease progression [HR = 0.447; 95% confidence interval (CI), 0.333-0.600; P < 0.001] and death (HR = 0.473; 95% CI, 0.320-0.700; P < 0.001). Of note, patients who achieved undetectable MRD early still benefitted from ASCT, with a remarkable improvement in the median MRD-negative duration (33.5-58.0 months, P < 0.001), progression-free survival (PFS; 46.0-88.3 months, P < 0.001), and overall survival (OS; 76.4 months to not reached, P = 0.003). These clinical benefits were more pronounced in patients with aggressive features (high-risk cytogenetic abnormalities or high tumor burden) compared with standard-risk patients. Similar results were observed in patients with detectable MRD after induction treatment. In addition, we identified four MRD-status transformation patterns following ASCT, which were strongly correlated with diverse survival outcomes (P < 0.001). Our study revealed the enhanced clinical significance of ASCT in patients with transplant-eligible myeloma, regardless of early MRD status, particularly for high-risk patients. Subsequent prospective trials are essential to validate these observations. Significance Achieving and maintaining undetectable MRD is the current treatment goal for multiple myeloma. Our results emphasized the remarkable clinical benefit of ASCT on MRD-negative duration, PFS, and OS in patients with multiple myeloma regardless of early MRD status. These favorable impacts were more evident in patients with aggressive features. Importantly, dynamic MRD monitoring among ASCT could facilitate personalized stratification of therapeutic approaches.
Collapse
Affiliation(s)
- Jiahui Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Wenqiang Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Huishou Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Jingyu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Lingna Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Xuehan Mao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Yuting Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Kenneth C. Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
- Tianjin Institutes of Health Science, Tianjin, P.R. China
| |
Collapse
|
204
|
Kraeber-Bodéré F, Jamet B, Bezzi D, Zamagni E, Moreau P, Nanni C. New Developments in Myeloma Treatment and Response Assessment. J Nucl Med 2023; 64:1331-1343. [PMID: 37591548 PMCID: PMC10478822 DOI: 10.2967/jnumed.122.264972] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/06/2023] [Indexed: 08/19/2023] Open
Abstract
Recent innovative strategies have dramatically redefined the therapeutic landscape for treating multiple myeloma patients. In particular, the development and application of immunotherapy and high-dose therapy have demonstrated high response rates and have prolonged remission duration. Over the past decade, new morphologic or hybrid imaging techniques have gradually replaced conventional skeletal surveys. PET/CT using 18F-FDG is a powerful imaging tool for the workup at diagnosis and for therapeutic evaluation allowing medullary and extramedullary assessment. The independent negative prognostic value for progression-free and overall survival derived from baseline PET-derived parameters such as the presence of extramedullary disease or paramedullary disease, as well as the number of focal bone lesions and SUVmax, has been reported in several large prospective studies. During therapeutic evaluation, 18F-FDG PET/CT is considered the reference imaging technique because it can be performed much earlier than MRI, which lacks specificity. Persistence of significant abnormal 18F-FDG uptake after therapy is an independent negative prognostic factor, and 18F-FDG PET/CT and medullary flow cytometry are complementary tools for detecting minimal residual disease before maintenance therapy. The definition of a PET metabolic complete response has recently been standardized and the interpretation criteria harmonized. The development of advanced PET analysis and radiomics using machine learning, as well as hybrid imaging with PET/MRI, offers new perspectives for multiple myeloma imaging. Most recently, innovative radiopharmaceuticals such as C-X-C chemokine receptor type 4-targeted small molecules and anti-CD38 radiolabeled antibodies have shown promising results for tumor phenotype imaging and as potential theranostics.
Collapse
Affiliation(s)
- Françoise Kraeber-Bodéré
- Médecine nucléaire, CHU Nantes, Nantes Université, Université Angers, INSERM, CNRS, CRCI2NA, F-44000, Nantes, France
| | - Bastien Jamet
- Médecine nucléaire, CHU Nantes, F-44000, Nantes, France
| | - Davide Bezzi
- Department of Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna. Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Philippe Moreau
- Hématologie, CHU Nantes, Nantes Université, Université Angers, INSERM, CNRS, CRCI2NA, F-44000, Nantes, France; and
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
205
|
Jew S, Bujarski S, Regidor B, Emamy-Sadr M, Swift R, Eades B, Kim S, Eshaghian S, Berenson JR. Clinical Outcomes and Serum B-Cell Maturation Antigen Levels in a Real-World Unselected Population of Newly Diagnosed Multiple Myeloma Patients. Target Oncol 2023; 18:735-747. [PMID: 37682503 DOI: 10.1007/s11523-023-00990-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Progression-free survival (PFS) and overall survival (OS) of newly diagnosed multiple myeloma (MM) patients have been widely published in the clinical trials setting, but data published from real-world settings are limited. OBJECTIVE We determined the survival and factors that predict outcomes among 161 unselected, newly diagnosed MM patients whose frontline therapy was started at a single clinic specializing in the treatment of this B-cell malignancy. PATIENTS AND METHODS None of these patients underwent an autologous stem cell transplantation as part of their initial therapy and the population had a high proportion (35%) of cytogenetic high-risk patients. RESULTS With a median follow-up of 42.7 months, the cohort had a median PFS of 22.8 months and a median OS of 136.2 months. The 1-, 3-, and 5-year survival rates were 97.5%, 85.3%, and 76.2%, respectively. These results are considerably better than those reported from patients enrolled in clinical trials and those from countries with national registries. Age <65 years predicted for a longer OS (p = 0.0004). Baseline serum B-cell maturation antigen (sBCMA) levels were also assessed and showed median and mean levels of 320.3 ng/mL and 551.1 ng/mL, respectively. Furthermore, patients with baseline sBCMA levels in the lowest quartile (≤136.2 ng/mL) showed a longer PFS (p = 0.0262). CONCLUSION These results provide clinicians with a real-world understanding of the survival of unselected, newly diagnosed patients initiating therapy in a clinic specializing in the care of MM patients.
Collapse
Affiliation(s)
- Scott Jew
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA
- Berenson Cancer Center, West Hollywood, CA, USA
| | - Sean Bujarski
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA
- Berenson Cancer Center, West Hollywood, CA, USA
| | | | | | | | | | | | | | - James R Berenson
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA.
- Berenson Cancer Center, West Hollywood, CA, USA.
- ONCOtherapeutics, West Hollywood, CA, USA.
- ONCOtracker, West Hollywood, CA, USA.
| |
Collapse
|
206
|
Kegyes D, Gulei D, Drula R, Cenariu D, Tigu B, Dima D, Tanase A, Badelita S, Buzoianu AD, Ciurea S, Ghiaur G, Terpos E, Ciechanover A, Einsele H, Tomuleasa C. Proteasome inhibition in combination with immunotherapies: State-of-the-Art in multiple myeloma. Blood Rev 2023; 61:101100. [PMID: 37291017 DOI: 10.1016/j.blre.2023.101100] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder accounting for around 1.8% of all neoplastic diseases. Nowadays, clinicians have a broad arsenal of drugs at their disposal for the treatment of MM, such as proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, bispecific antibodies, CAR T-cell therapies and antibody-drug conjugates. In this paper we briefly highlight essential clinical elements relating to proteasome inhibitors, such as bortezomib, carfilzomib and ixazomib. Studies suggest that the early use of immunotherapy may improve outcomes significantly. Therefore, in our review we specifically focus on the combination therapy of proteasome inhibitors with novel immunotherapies and/or transplant. A high number of patients develop PI resistance. Thus, we also review new generation PIs, such as marizomib, oprozomib (ONX0912) and delanzomib (CEP-18770) and their combinations with immunotherapies.
Collapse
Affiliation(s)
- David Kegyes
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Rares Drula
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Diana Cenariu
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bogdan Tigu
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Alina Tanase
- Department of Hematology and Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Sorina Badelita
- Department of Hematology and Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Anca-Dana Buzoianu
- Department of Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Stefan Ciurea
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California Irvine, CA, United States
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Aaron Ciechanover
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hermann Einsele
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
| |
Collapse
|
207
|
Ramasamy K, Avet-Loiseau H, Hveding Blimark C, Delforge M, Gay F, Manier S, Martinez-Lopez J, Mateos MV, Mohty M, van de Donk NW, Weisel K. Measurable Residual Disease Testing in Multiple Myeloma Routine Clinical Practice: A Modified Delphi Study. Hemasphere 2023; 7:e942. [PMID: 37663672 PMCID: PMC10470794 DOI: 10.1097/hs9.0000000000000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
We used a modified Delphi approach to establish areas of consensus and nonconsensus regarding the utility of determining measurable residual disease (MRD) to assess multiple myeloma (MM) treatment response, which may inform disease management and design of future clinical trials. This modified Delphi study incorporated 2 iterative rounds of surveys to evaluate the opinions of an expert panel of 61 practicing hematological oncologists from across 14 countries in Europe concerning the use of MRD testing in MM management. Survey 1 assessed experts' opinions on MRD testing in different clinical situations and associated challenges. Survey 2 focused on the lack of consensus areas identified in survey 1. Consensus to an individual question was defined a priori as 75% agreement or disagreement by the panel. From the 2 rounds of surveys, the experts reached consensus agreement that MRD testing should be performed in newly diagnosed or relapsed patients who achieved complete response (CR) or better after transplantation. In transplant-ineligible patients, experts recommended MRD testing in those who are ≤70 years old and in CR. If a patient was previously positive on positron-emission tomography and computed tomography (PET/CT), both MRD and PET/CT should be assessed at CR. MRD testing should be performed ≤6 months after transplantation and every 6-12 months in continuously treated patients in CR. There was no consensus on making treatment decisions based on MRD status. MRD testing is an important component of clinical management in MM. Additional data will further clarify the role of MRD in guiding treatment decisions.
Collapse
Affiliation(s)
- Karthik Ramasamy
- Oxford University Hospitals NHS Foundation Trust, Radcliffe Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Hervé Avet-Loiseau
- University Institute of Cancer Toulouse, University Hospital of Toulouse, Toulouse, France
| | | | | | | | | | | | - Maria Victoria Mateos
- University Hospital of Salamanca, Salamanca Biomedical Research Institute (IBSAL), CIC, Ciberonc, Salamanca, Spain
| | - Mohamad Mohty
- Hospital Saint-Antoine, Sorbonne University, INSERM UMRs 938, Paris, France
| | | | - Katja Weisel
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
208
|
Liccardo F, Śniegocka M, Tito C, Iaiza A, Ottone T, Divona M, Travaglini S, Mattei M, Cicconi R, Miglietta S, Familiari G, Nottola SA, Petrozza V, Tamagnone L, Voso MT, Masciarelli S, Fazi F. Retinoic acid and proteotoxic stress induce AML cell death overcoming stromal cell protection. J Exp Clin Cancer Res 2023; 42:223. [PMID: 37653435 PMCID: PMC10469880 DOI: 10.1186/s13046-023-02793-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) patients bearing the ITD mutation in the tyrosine kinase receptor FLT3 (FLT3-ITD) present a poor prognosis and a high risk of relapse. FLT3-ITD is retained in the endoplasmic reticulum (ER) and generates intrinsic proteotoxic stress. We devised a strategy based on proteotoxic stress, generated by the combination of low doses of the differentiating agent retinoic acid (R), the proteasome inhibitor bortezomib (B), and the oxidative stress inducer arsenic trioxide (A). METHODS We treated FLT3-ITD+ AML cells with low doses of the aforementioned drugs, used alone or in combinations and we investigated the induction of ER and oxidative stress. We then performed the same experiments in an in vitro co-culture system of FLT3-ITD+ AML cells and bone marrow stromal cells (BMSCs) to assess the protective role of the niche on AML blasts. Eventually, we tested the combination of drugs in an orthotopic murine model of human AML. RESULTS The combination RBA exerts strong cytotoxic activity on FLT3-ITD+ AML cell lines and primary blasts isolated from patients, due to ER homeostasis imbalance and generation of oxidative stress. AML cells become completely resistant to the combination RBA when treated in co-culture with BMSCs. Nonetheless, we could overcome such protective effects by using high doses of ascorbic acid (Vitamin C) as an adjuvant. Importantly, the combination RBA plus ascorbic acid significantly prolongs the life span of a murine model of human FLT3-ITD+ AML without toxic effects. Furthermore, we show for the first time that the cross-talk between AML and BMSCs upon treatment involves disruption of the actin cytoskeleton and the actin cap, increased thickness of the nuclei, and relocalization of the transcriptional co-regulator YAP in the cytosol of the BMSCs. CONCLUSIONS Our findings strengthen our previous work indicating induction of proteotoxic stress as a possible strategy in FLT3-ITD+ AML therapy and open to the possibility of identifying new therapeutic targets in the crosstalk between AML and BMSCs, involving mechanotransduction and YAP signaling.
Collapse
Affiliation(s)
- Francesca Liccardo
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Martyna Śniegocka
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Claudia Tito
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Alessia Iaiza
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Serena Travaglini
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Maurizio Mattei
- Department of Biology, University of Tor Vergata, Rome, Italy
- Centro Interdipartimentale-CIMETA, University of Tor Vergata, Rome, Italy
| | - Rosella Cicconi
- Centro Interdipartimentale-CIMETA, University of Tor Vergata, Rome, Italy
| | - Selenia Miglietta
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Section of Human Anatomy, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Familiari
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Section of Human Anatomy, Sapienza University of Rome, Rome, Italy
| | - Stefania Annarita Nottola
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Section of Human Anatomy, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Petrozza
- Department of Medico-Surgical Sciences & Biotechnologies, Center for Biophotonics, Sapienza University of Rome, Latina, Italy
| | - Luca Tamagnone
- Department of Life Sciences and Public Health, Histology and Embryology Unit, Catholic University of the Sacred Hearth, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - Silvia Masciarelli
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy.
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
209
|
Charalampous C, Goel U, Kapoor P, Binder M, Buadi FK, Cook J, Dingli D, Dispenzieri A, Fonder AL, Gertz MA, Gonsalves W, Hayman SR, Hobbs MA, Hwa YL, Kourelis T, Lacy MQ, Leung N, Lin Y, Warsame R, Kyle RA, Rajkumar SV, Kumar SK. Outcomes of patients with primary refractory multiple myeloma in the era of triplet and quadruplet induction therapy. Blood Adv 2023; 7:4371-4380. [PMID: 37603349 PMCID: PMC10432608 DOI: 10.1182/bloodadvances.2023009681] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Patients with multiple myeloma (MM) who do not respond to initial therapy have worse outcomes than primary responders, and effective treatments are lacking in this population. However, the outcomes of primary refractory disease in the modern treatment era have not yet been studied. We reviewed patients with MM treated with triplet/quadruplet therapy at our institution to assess the incidence of primary refractory disease and the impact of salvage therapies in this population. We identified 1127 patients, of whom 1086 were evaluated for hematologic responses after 4 to 6 cycles. Of these, 93.3% (1013) had evidence of response, whereas 6.7% (73) had primary refractory disease. With a median overall survival (OS) of 51.3 months, patients with primary refractory disease had an increased risk of shorter survival in univariable and multivariable analyses (hazard ratio [HR], 3.5 [95% confidence interval (CI), 2.5-4.9]; HR, 4.3 [95% CI, 2.6-6.9], respectively). In the subgroup analysis of patients with primary refractory disease, those who received second-line autologous stem cell transplantation (ASCT) had increased second progression-free survival (20.9 vs 8.1 months; P < .01) and second OS (74.7 vs 31.3 months; P = .02) compared with patients who did not. We conclude that early progression remains a significant factor for shorter OS in the current era, and salvage ASCT could be the most beneficial option for this population.
Collapse
Affiliation(s)
| | - Utkarsh Goel
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Prashant Kapoor
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Moritz Binder
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Francis K. Buadi
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Joselle Cook
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - David Dingli
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Angela Dispenzieri
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Amie L. Fonder
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Morie A. Gertz
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Wilson Gonsalves
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Suzanne R. Hayman
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Miriam A. Hobbs
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Yi L. Hwa
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Taxiarchis Kourelis
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Martha Q. Lacy
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Nelson Leung
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Rahma Warsame
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Robert A. Kyle
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - S. Vincent Rajkumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Shaji K. Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
210
|
Petrilla C, Galloway J, Kudalkar R, Ismael A, Cottini F. Understanding DNA Damage Response and DNA Repair in Multiple Myeloma. Cancers (Basel) 2023; 15:4155. [PMID: 37627183 PMCID: PMC10453069 DOI: 10.3390/cancers15164155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy characterized by several genetic abnormalities, including chromosomal translocations, genomic deletions and gains, and point mutations. DNA damage response (DDR) and DNA repair mechanisms are altered in MM to allow for tumor development, progression, and resistance to therapies. Damaged DNA rarely induces an apoptotic response, given the presence of ataxia-telangiectasia mutated (ATM) loss-of-function or mutations, as well as deletions, mutations, or downregulation of tumor protein p53 (TP53) and tumor protein p73 (TP73). Moreover, DNA repair mechanisms are either hyperactive or defective to allow for rapid correction of the damage or permissive survival. Medications used to treat patients with MM can induce DNA damage, by either direct effects (mono-adducts induced by melphalan), or as a result of reactive oxygen species (ROS) production by proteasome inhibitors such as bortezomib. In this review, we will describe the mechanisms of DDR and DNA repair in normal tissues, the contribution of these pathways to MM disease progression and other phenotypes, and the potential therapeutic opportunities for patients with MM.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Cottini
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
211
|
Mo CC, Yee AJ, Midha S, Hartley‐Brown MA, Nadeem O, O'Donnell EK, Bianchi G, Sperling AS, Laubach JP, Richardson PG. Selinexor: Targeting a novel pathway in multiple myeloma. EJHAEM 2023; 4:792-810. [PMID: 37601856 PMCID: PMC10435704 DOI: 10.1002/jha2.709] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 08/22/2023]
Abstract
Selinexor is an orally bioavailable selective inhibitor of nuclear export compound that inhibits exportin-1 (XPO1), a novel therapeutic target that is overexpressed in multiple myeloma (MM) and is responsible for the transport of ∼220 nuclear proteins to the cytoplasm, including tumour suppressor proteins. Inhibition of this process has demonstrated substantial antimyeloma activity in preclinical studies, both alone and in combination with established MM therapeutics. Based on a clinical trial programme encompassing multiple combination regimens, selinexor-based therapy has been approved for the treatment of relapsed/refractory MM (RRMM), with selinexor-dexamethasone approved in the later-relapse setting for penta-refractory patients and selinexor-bortezomib-dexamethasone approved for patients who have received ≥1 prior therapy. Here, we provide a comprehensive review of the clinical data on selinexor-based regimens, including recent updates from the 2022 American Society of Hematology annual meeting, and summarise ongoing studies of this novel targeted agent in newly diagnosed MM and RRMM.
Collapse
Affiliation(s)
- Clifton C. Mo
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
| | - Andrew J. Yee
- Massachusetts General Cancer CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Shonali Midha
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
- Division of HematologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Monique A. Hartley‐Brown
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
- Division of HematologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Omar Nadeem
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
| | - Elizabeth K. O'Donnell
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
- Division of HematologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Giada Bianchi
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
- Division of HematologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Adam S. Sperling
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
- Division of HematologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Jacob P. Laubach
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
| | - Paul G. Richardson
- Department of Medical OncologyDana‐Farber Cancer InstituteJerome Lipper Center for Multiple Myeloma ResearchHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
212
|
Pasvolsky O, Marcoux C, Milton DR, Tanner MR, Bashir Q, Srour S, Saini N, Lin P, Ramdial J, Nieto Y, Lee HC, Patel KK, Kebriaei P, Tewari P, Crawford-Suber L, Thomas SK, Weber DM, Orlowski RZ, Shpall EJ, Champlin RE, Qazilbash MH. Outcomes of young adults (aged ≤ 40 years) with newly diagnosed multiple myeloma after up-front autologous stem cell transplant. Br J Haematol 2023; 202:866-873. [PMID: 37376789 PMCID: PMC10527395 DOI: 10.1111/bjh.18944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Multiple myeloma (MM) primarily affects older patients. There are scarce data on the outcomes of young adults undergoing autologous transplantation (auto-HCT). In this single-centre analysis, we included 117 younger patients, with a median age of 37 years (range 22-40) at transplant. Seventeen (15%) patients had high-risk cytogenetics. Before transplant, 10% of patients achieved ≥CR and 44% achieved ≥VGPR. At best post-transplant response, 56% and 77% of patients achieved ≥CR and ≥VGPR respectively. With a median follow-up for survivors of 72.6 months (range 0.9-238.0), median PFS and OS were 43.1 months (95% CI 31.2-65.0) and 146.6 months (95% CI 100.0-208.1) respectively. Patients who underwent auto-HCT after 2010 had better median PFS (84.9 months vs. 28.2 months, p < 0.001) and OS (NR vs. 91.8 months, p < 0.001) compared with those transplanted earlier. In multi-variate analysis, achieving ≥CR as best post-transplant response was associated with improved PFS (HR [95% CI] 0.55 [0.32-0.95], p = 0.032), while achieving ≥VGPR was predictive of superior OS (0.32 [0.16-0.62], p < 0.001). Three patients (3%) developed a second primary malignancy. Younger MM patients had durable survival after auto-HCT, which further improved after the availability of novel anti-myeloma drugs in recent years. Depth of response following transplant remains a key predictor of survival.
Collapse
Affiliation(s)
- Oren Pasvolsky
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Curtis Marcoux
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Denái R. Milton
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark R. Tanner
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeraj Saini
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hans C. Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krina K. Patel
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Priti Tewari
- Department of Pediatrics Patient Care, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lindsay Crawford-Suber
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Sheeba K. Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Donna M. Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert Z. Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E. Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muzaffar H. Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
213
|
Tan CR, Derkach A, Nemirovsky D, Ciardiello A, Diamond B, Hultcrantz M, Hassoun H, Mailankody S, Shah U, Maclachlan K, Patel D, Lahoud OB, Landau HJ, Chung DJ, Shah GL, Scordo M, Giralt SA, Lesokhin A, Usmani SZ, Landgren O, Korde N. Bortezomib, lenalidomide and dexamethasone (VRd) vs carfilzomib, lenalidomide and dexamethasone (KRd) as induction therapy in newly diagnosed multiple myeloma. Blood Cancer J 2023; 13:112. [PMID: 37491332 PMCID: PMC10368661 DOI: 10.1038/s41408-023-00882-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/17/2023] [Accepted: 07/11/2023] [Indexed: 07/27/2023] Open
Abstract
Lenalidomide and dexamethasone with bortezomib (VRd) or carfilzomib (KRd) are commonly used induction regimens in the U.S. This single-center, retrospective study evaluated outcomes and safety of VRd and KRd. Primary endpoint was progression-free survival (PFS). Of 389 patients with newly diagnosed multiple myeloma, 198 received VRd and 191 received KRd. Median PFS was not reached (NR) in both groups; 5-year PFS was 56% (95%CI, 48-64%) for VRd and 67% (60-75%) for KRd (P = 0.027). Estimated 5-year EFS was 34% (95%CI, 27-42%) for VRd and 52% (45-60%) for KRd (P < 0.001) with corresponding 5-year OS of 80% (95%CI, 75-87%) and 90% (85-95%), respectively (P = 0.053). For standard-risk patients, 5-year PFS was 68% (95%CI, 60-78%) for VRd and 75% (65-85%) for KRd (P = 0.20) with 5-year OS of 87% (95%CI, 81-94%) and 93% (87-99%), respectively (P = 0.13). For high-risk patients, median PFS was 41 months (95%CI, 32.8-61.1) for VRd and 70.9 months (58.2-NR) for KRd (P = 0.016). Respective 5-year PFS and OS were 35% (95%CI, 24-51%) and 69% (58-82%) for VRd and 58% (47-71%) and 88% (80-97%, P = 0.044) for KRd. Overall, KRd resulted in improved PFS and EFS with a trend toward improved OS compared to VRd with associations primarily driven by improvements in outcome for high-risk patients.
Collapse
Affiliation(s)
- Carlyn Rose Tan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Andriy Derkach
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Nemirovsky
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amanda Ciardiello
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Diamond
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Urvi Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dhwani Patel
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oscar B Lahoud
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heather J Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David J Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Saad Z Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
214
|
Bao A, Zhao Q, Kudalkar R, Rodriguez J, Sharma N, Bumma N, Devarakonda SS, Khan AM, Umyarova E, Rosko AE, Benson D, Cottini F. Impact of interval progression before autologous stem cell transplant in patients with multiple myeloma. Front Oncol 2023; 13:1216461. [PMID: 37554170 PMCID: PMC10405820 DOI: 10.3389/fonc.2023.1216461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/28/2023] [Indexed: 08/10/2023] Open
Abstract
In transplant-eligible patients who undergo upfront autologous stem cell transplant (ASCT) for multiple myeloma (MM), standard practice is to treat with six to eight cycles of induction therapy followed by high-dose chemotherapy with ASCT. A gap between the end of induction and the day of ASCT exists to allow stem cell mobilization and collection. Despite attempts to limit the length of this interval, we noticed that some patients experience interval progression (IP) of disease between the end of induction therapy and the day of ASCT. We analyzed 408 MM patients who underwent ASCT between 2011 and 2016. The median length of the interval between end of induction and ASCT was 38 days. We observed that 26% of patients in the entire cohort and 23.6% of patients who received induction with bortezomib-lenalidomide-dexamethasone (VRD) experienced IP. These patients deepened their responses with ASCT, independently of induction regimen. In the entire cohort, IP was significantly associated with shorter PFS in the univariable analysis (Hazard Ratio, HR = 1.37, P = 0.022) but not in the multivariable analysis (HR = 1.14, P = 0.44). However, analyzing only patients who received VRD as induction, progression-free survival (PFS) remained inferior in both the univariable (HR = 2.02; P = 0.002) and the multivariable analyses (HR = 1.96; P = 0.01). T cells and natural killer (NK) cells are increasingly studied targets of immunomodulatory therapy, as immune dysfunction is known to occur in patients with MM. Peripheral blood from 35 MM patients were analyzed. At time of ASCT, patients with IP had significantly increased percentages of CD3+CD8+CD57+ CD28- (P = 0.05) and CD3+CD4+LAG3+ (P = 0.0022) T-cells, as well as less CD56bright and CD56dim NK cells bearing activated markers such as CD69, NKG2D, and CD226. These data suggest that IP can impact the length of response to ASCT; therefore, further studies on the management of these patients are needed.
Collapse
Affiliation(s)
- Alicia Bao
- The Ohio State University, College of Medicine, Columbus, OH, United States
| | - Qiuhong Zhao
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ruchi Kudalkar
- The Ohio State University, College of Medicine, Columbus, OH, United States
| | - Jose Rodriguez
- The Ohio State University, College of Medicine, Columbus, OH, United States
- School of Medicine, Ponce Health Science University, Ponce, Puerto Rico
| | - Nidhi Sharma
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Naresh Bumma
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Srinivas S. Devarakonda
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Abdullah M. Khan
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Elvira Umyarova
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ashley E. Rosko
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Don Benson
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Francesca Cottini
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
215
|
Bumma N, Dhakal B, Fraser R, Estrada-Merly N, Anderson K, Freytes CO, Hildebrandt GC, Holmberg L, Krem MM, Lee C, Lekakis L, Lazarus HM, Mian H, Murthy HS, Nathan S, Nishihori T, Parrondo R, Patel SS, Solh M, Strouse C, Vesole DH, Kumar S, Qazilbash MH, Shah N, D’Souza A, Sidana S. Impact of bortezomib-based versus lenalidomide maintenance therapy on outcomes of patients with high-risk multiple myeloma. Cancer 2023; 129:2179-2191. [PMID: 37021929 PMCID: PMC10516285 DOI: 10.1002/cncr.34778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/16/2023] [Accepted: 02/02/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Lenalidomide maintenance after autologous stem cell transplant (ASCT) in multiple myeloma (MM) results in superior progression-free survival and overall survival. However, patients with high-risk multiple myeloma (HRMM) do not derive the same survival benefit from lenalidomide maintenance compared with standard-risk patients. The authors sought to determine the outcomes of bortezomib-based maintenance compared with lenalidomide maintenance in patients with HRMM undergoing ASCT. METHODS In total, the authors identified 503 patients with HRMM who were undergoing ASCT within 12 months of diagnosis from January 2013 to December 2018 after receiving triplet novel-agent induction in the Center for International Blood and Marrow Transplant Research database. HRMM was defined as deletion 17p, t(14;16), t(4;14), t(14;20), or chromosome 1q gain. RESULTS Three hundred fifty-seven patients (67%) received lenalidomide alone, and 146 (33%) received bortezomib-based maintenance (with bortezomib alone in 58%). Patients in the bortezomib-based maintenance group were more likely to harbor two or more high-risk abnormalities and International Staging System stage III disease (30% vs. 22%; p = .01) compared with the lenalidomide group (24% vs. 15%; p < .01). Patients who were receiving lenalidomide maintenance had superior progression-free survival at 2 years compared with those who were receiving either bortezomib monotherapy or combination therapy (75% vs. 63%; p = .009). Overall survival at 2 years was also superior in the lenalidomide group (93% vs. 84%; p = .001). CONCLUSIONS No superior outcomes were observed in patients with HRMM who received bortezomib monotherapy or (to a lesser extent) in those who received bortezomib in combination as maintenance compared with lenalidomide alone. Until prospective data from randomized clinical trials are available, post-transplant therapy should be tailored to each patient with consideration for treating patients in clinical trials that target novel therapeutic strategies for HRMM, and lenalidomide should remain a cornerstone of treatment.
Collapse
Affiliation(s)
- Naresh Bumma
- James Cancer Center, Ohio State Medical Center, Columbus, Ohio, USA
| | - Binod Dhakal
- Bone Marrow Transplant and Cellular Therapy Program, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Raphael Fraser
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for International Blood and Marrow Transplant Research (CIBMTR), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Noel Estrada-Merly
- Center for International Blood and Marrow Transplant Research (CIBMTR), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - César O. Freytes
- Bone Marrow Transplant Program, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | - Leona Holmberg
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Maxwell M. Krem
- Division of Hematology/BMT, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, USA
| | - Cindy Lee
- Department of Hematology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Lazaros Lekakis
- Division of Transplantation and Cellular Therapy, University of Miami Hospital and Clinics, Sylvester Comprehensive Cancer Center, Miami, Florida, USA
| | - Hillard M. Lazarus
- University Hospital Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hira Mian
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Hemant S. Murthy
- Blood and Marrow Transplantation Program, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Sunita Nathan
- Section of Bone Marrow Transplant and Cell Therapy, Division of Hematology, Oncology, and Cell Therapy, Rush University Medical Center, Chicago, Illinois, USA
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Ricardo Parrondo
- Blood and Marrow Transplantation Program, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Sagar S. Patel
- Transplant and Cellular Therapy Program, Huntsman Cancer Center Institute, University of Utah, Salt Lake City, Utah, USA
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia, USA
| | - Christopher Strouse
- Division of Hematology, Oncology, and Bone Marrow Transplantation, University of Iowa, Iowa City, Iowa, USA
| | - David H. Vesole
- Jonn Theurer Cancer Center at Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Shaji Kumar
- Hematology/Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Muzaffar H. Qazilbash
- Department of Blood and Marrow Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nina Shah
- Division of Hematology-Oncology, University of California San Francisco, San Francisco, California, USA
| | - Anita D’Souza
- Center for International Blood and Marrow Transplant Research (CIBMTR), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford Health Care, Stanford, California, USA
| |
Collapse
|
216
|
Geyer MB, Shaffer BC, Bhatnagar B, Mims AS, Klein V, Dilip D, Glass JL, Lozanski G, Hassoun H, Landau H, Zhang Y, Xiao W, Roshal M, Park JH. Lenalidomide-associated B-cell ALL: clinical and pathologic correlates and sensitivity to lenalidomide withdrawal. Blood Adv 2023; 7:3087-3098. [PMID: 36827680 PMCID: PMC10362546 DOI: 10.1182/bloodadvances.2022009212] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/31/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Lenalidomide is an effective component of induction and maintenance therapy for multiple myeloma, though with a risk of secondary malignancies, including acute lymphoblastic leukemia (ALL). In contrast to therapy-related myeloid neoplasia, lenalidomide-associated lymphoblastic neoplasia remains poorly characterized. We conducted a dual institution retrospective study of 32 ALL cases that arose after lenalidomide maintenance (all B-lineage, 31/32 BCR::ABL-negative). B-cell ALL (B-ALL) was diagnosed at median 54 months (range, 5-119) after first exposure to lenalidomide and after median 42 months of cumulative lenalidomide exposure (range, 2-114). High incidence of TP53 mutations (9/19 evaluable cases) and low hypodiploidy (8/26 patients) were identified. Despite median age of 65 years and poor-risk B-ALL features observed in the cohort, rates of complete response (CR) or CR with incomplete hematologic recovery were high (25/28 patients receiving treatment). Median event-free survival was 35.4 months among treated patients (not reached among those undergoing allogeneic hematopoietic cell transplantation [HCT]). Sixteen patients remain alive without evidence of B-ALL after HCT or extended maintenance therapy. We also describe regression of B-ALL or immature B-cell populations with B-ALL immunophenotype after lenalidomide discontinuation in 5 patients, suggesting lenalidomide may drive leukemic progression even after initiation of lymphoblastic neoplasia and that lenalidomide withdrawal alone may be an appropriate first-line intervention in selected patients. Monitoring for early B-ALL-like proliferations may offer opportunities for lenalidomide withdrawal to prevent progression. Established combination chemotherapy regimens, newer surface antigen-targeted approaches, and allogeneic HCT are effective in many patients with lenalidomide-associated B-ALL and should be offered to medically fit patients.
Collapse
Affiliation(s)
- Mark B. Geyer
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cell Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brian C. Shaffer
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bhavana Bhatnagar
- Section of Hematology/Oncology, Department of Medicine, West Virginia University, West Virginia University Cancer Institute, Morgantown, WV
| | - Alice S. Mims
- Acute Leukemia Program, The Ohio State University, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Victoria Klein
- Acute Leukemia Program, The Ohio State University, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Deepika Dilip
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jacob L. Glass
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gerard Lozanski
- Department of Pathology, The Ohio State University, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heather Landau
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wenbin Xiao
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mikhail Roshal
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jae H. Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cell Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
217
|
Cowan AJ, Pont MJ, Sather BD, Turtle CJ, Till BG, Libby EN, Coffey DG, Tuazon SA, Wood B, Gooley T, Wu VQ, Voutsinas J, Song X, Shadman M, Gauthier J, Chapuis AG, Milano F, Maloney DG, Riddell SR, Green DJ. γ-Secretase inhibitor in combination with BCMA chimeric antigen receptor T-cell immunotherapy for individuals with relapsed or refractory multiple myeloma: a phase 1, first-in-human trial. Lancet Oncol 2023; 24:811-822. [PMID: 37414012 PMCID: PMC10783021 DOI: 10.1016/s1470-2045(23)00246-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND γ-Secretase inhibitors (GSIs) increase B cell maturation antigen (BCMA) density on malignant plasma cells and enhance antitumour activity of BCMA chimeric antigen receptor (CAR) T cells in preclinical models. We aimed to evaluate the safety and identify the recommended phase 2 dose of BCMA CAR T cells in combination with crenigacestat (LY3039478) for individuals with relapsed or refractory multiple myeloma. METHODS We conducted a phase 1, first-in-human trial combining crenigacestat with BCMA CAR T-cells at a single cancer centre in Seattle, WA, USA. We included individuals aged 21 years or older with relapsed or refractory multiple myeloma, previous autologous stem-cell transplant or persistent disease after more than four cycles of induction therapy, and Eastern Cooperative Oncology Group performance status of 0-2, regardless of previous BCMA-targeted therapy. To assess the effect of the GSI on BCMA surface density on bone marrow plasma cells, participants received GSI during a pretreatment run-in, consisting of three doses administered 48 h apart. BCMA CAR T cells were infused at doses of 50 × 106 CAR T cells, 150 × 106 CAR T cells, 300 × 106 CAR T cells, and 450 × 106 CAR T cells (total cell dose), in combination with the 25 mg crenigacestat dosed three times a week for up to nine doses. The primary endpoints were the safety and recommended phase 2 dose of BCMA CAR T cells in combination with crenigacestat, an oral GSI. This study is registered with ClinicalTrials.gov, NCT03502577, and has met accrual goals. FINDINGS 19 participants were enrolled between June 1, 2018, and March 1, 2021, and one participant did not proceed with BCMA CAR T-cell infusion. 18 participants (eight [44%] men and ten [56%] women) with multiple myeloma received treatment between July 11, 2018, and April 14, 2021, with a median follow up of 36 months (95% CI 26 to not reached). The most common non-haematological adverse events of grade 3 or higher were hypophosphataemia in 14 (78%) participants, fatigue in 11 (61%), hypocalcaemia in nine (50%), and hypertension in seven (39%). Two deaths reported outside of the 28-day adverse event collection window were related to treatment. Participants were treated at doses up to 450 × 106 CAR+ cells, and the recommended phase 2 dose was not reached. INTERPRETATIONS Combining a GSI with BCMA CAR T cells appears to be well tolerated, and crenigacestat increases target antigen density. Deep responses were observed among heavily pretreated participants with multiple myeloma who had previously received BCMA-targeted therapy and those who were naive to previous BCMA-targeted therapy. Further study of GSIs given with BCMA-targeted therapeutics is warranted in clinical trials. FUNDING Juno Therapeutics-a Bristol Myers Squibb company and the National Institutes of Health.
Collapse
Affiliation(s)
- Andrew J Cowan
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Margot J Pont
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | | | - Cameron J Turtle
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Brian G Till
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Edward N Libby
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - David G Coffey
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Sherilyn A Tuazon
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Brent Wood
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Ted Gooley
- Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Vicky Q Wu
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA; Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jenna Voutsinas
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA; Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Xiaoling Song
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA; Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Mazyar Shadman
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jordan Gauthier
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Aude G Chapuis
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Filippo Milano
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - David G Maloney
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Stanley R Riddell
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Damian J Green
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA.
| |
Collapse
|
218
|
Sharma N, Benson E, Zhao Q, Nunnelee J, Cottini F, Elder P, Rosko A, Bumma N, Khan A, Umyarova E, Devarakonda S, Efebera YA, Benson DM. Survival outcomes following autologous stem cell transplant with melphalan 140mg/m 2 versus 200mg/m 2 preparative regimens in patients with multiple myeloma. Leuk Lymphoma 2023; 64:1315-1321. [PMID: 37199099 DOI: 10.1080/10428194.2023.2213366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
The standard preparative regimen for autologous stem cell transplant (ASCT) in multiple myeloma (MM) is 200 mg/m2 of intravenous melphalan; however, a dose of 140 mg/m2 is often used when concerns exist related to patient age, performance status, organ function, and other factors. It is unclear whether a lower dose of melphalan impacts post-transplant survival outcomes. We performed a retrospective review of 930 patients with MM who underwent ASCT with 200 mg/m2 versus 140 mg/m2 melphalan. On univariable analysis, no difference in progression-free survival (PFS) was observed, however, an overall survival (OS) benefit was observed in patients receiving 200 mg/m2 melphalan (p = 0.04). Multivariable analyses showed patients receiving 140 mg/m2 faired no worse than those receiving 200 mg/m2. While a subset of younger patients with normal renal function may achieve superior OS with a standard dose of 200 mg/m2 melphalan, these findings suggest an opportunity to individualize the ASCT preparative regimen to optimize outcomes.
Collapse
Affiliation(s)
- Nidhi Sharma
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Evan Benson
- Pre-Medical Program, University of Dayton, Dayton, OH, USA
| | - Qiuhong Zhao
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Jordan Nunnelee
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Francesca Cottini
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Patrick Elder
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ashley Rosko
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Naresh Bumma
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Abdullah Khan
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Elvira Umyarova
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Srinivas Devarakonda
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Yvonne A Efebera
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Don M Benson
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- Bone Marrow Transplantation & Cellular Therapy, OhioHealth, Columbus, OH, USA
| |
Collapse
|
219
|
Hashmi H, Davis JA, Abdallah AO. 'A stitch in time saves nine': early stem cell transplant continues to improve outcomes in patients with newly diagnosed multiple myeloma. Expert Rev Hematol 2023; 16:561-563. [PMID: 37334674 DOI: 10.1080/17474086.2023.2227789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/16/2023] [Indexed: 06/20/2023]
Affiliation(s)
- Hamza Hashmi
- Division of Hematology and Medical Oncology, Medical University of South Carolina
| | - James A Davis
- Division of Hematology and Medical Oncology, Medical University of South Carolina
| | - Al-Ola Abdallah
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center
| |
Collapse
|
220
|
Oriol A, Abril L, Ibarra G. First-line treatment of multiple myeloma in both transplant and non-transplant candidates. Expert Rev Anticancer Ther 2023; 23:685-698. [PMID: 37194283 DOI: 10.1080/14737140.2023.2213891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023]
Abstract
INTRODUCTION The last decade's progress in the treatment of newly diagnosed multiple myeloma has relied on the synergistic combination of agents with different mechanisms of action, basically proteasome inhibitors, immunomodulatory agents, and monoclonal antibodies, in order to achieve the deepest possible response early in the course of treatment. Following induction, several therapeutic strategies aim to improve and maintain response. AREAS COVERED The manuscript reviews available data for the treatment of newly diagnosed multiple myeloma patients with a focus on most recent induction and maintenance combinations and the still important role of autologous stem transplantation. Future perspectives in the light of initial results from ongoing clinical trials are also addressed. EXPERT OPINION Remarkable progress has been made in myeloma treatment due to the integration of immunomodulators, proteasome inhibitors, monoclonal antibodies, and high dose therapy in the frontline setting. Upfront therapy may be further improved intensifying induction combinations, adapting high dose therapy and consolidation strategies to the patient's profile, improving maintenance in high-risk individuals, or limiting maintenance duration in those with a better prognosis. Evidence needs to be reviewed, taking into account the therapeutic objectives at each treatment stage and patient specific risk factors.
Collapse
Affiliation(s)
- Albert Oriol
- Institut Català d'Oncologia and Institut Josep Carreras. Hospital Germans Trias i Pujol, Carretera del Canyet, Barcelona, Spain
| | - Laura Abril
- Institut Català d'Oncologia and Institut Josep Carreras. Hospital Germans Trias i Pujol, Carretera del Canyet, Barcelona, Spain
| | - Gladys Ibarra
- Institut Català d'Oncologia and Institut Josep Carreras. Hospital Germans Trias i Pujol, Carretera del Canyet, Barcelona, Spain
| |
Collapse
|
221
|
Ragon BK, Shah MV, D’Souza A, Estrada-Merly N, Gowda L, George G, de Lima M, Hashmi S, Kharfan-Dabaja MA, Majhail NS, Banerjee R, Saad A, Hildebrandt GC, Mian H, Abid MB, Battiwalla M, Lekakis LJ, Patel SS, Murthy HS, Nieto Y, Strouse C, Badawy SM, Al Hadidi S, Dholaria B, Aljurf M, Vesole DH, Lee CH, Pawarode A, Gergis U, Miller KC, Holmberg LA, Afrough A, Solh M, Munshi PN, Nishihori T, Anderson LD, Wirk B, Kaur G, Qazilbash MH, Shah N, Kumar SK, Usmani SZ. Impact of second primary malignancy post-autologous transplantation on outcomes of multiple myeloma: a CIBMTR analysis. Blood Adv 2023; 7:2746-2757. [PMID: 36827681 PMCID: PMC10275699 DOI: 10.1182/bloodadvances.2022009138] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
The overall survival (OS) has improved significantly in multiple myeloma (MM) over the last decade with the use of proteasome inhibitor and immunomodulatory drug-based combinations, followed by high-dose melphalan and autologous hematopoietic stem cell transplantation (auto-HSCT) and subsequent maintenance therapies in eligible newly diagnosed patients. However, clinical trials using auto-HSCT followed by lenalidomide maintenance have shown an increased risk of second primary malignancies (SPM), including second hematological malignancies (SHM). We evaluated the impact of SPM and SHM on progression-free survival (PFS) and OS in patients with MM after auto-HSCT using CIBMTR registry data. Adult patients with MM who underwent first auto-HSCT in the United States with melphalan conditioning regimen from 2011 to 2018 and received maintenance therapy were included (n = 3948). At a median follow-up of 37 months, 175 (4%) patients developed SPM, including 112 (64%) solid, 36 (20%) myeloid, 24 (14%) SHM, not otherwise specified, and 3 (2%) lymphoid malignancies. Multivariate analysis demonstrated that SPM and SHM were associated with an inferior PFS (hazard ratio [HR] 2.62, P < .001 and HR 5.01, P < .001, respectively) and OS (HR 3.85, P < .001 and HR 8.13, P < .001, respectively). In patients who developed SPM and SHM, MM remained the most frequent primary cause of death (42% vs 30% and 53% vs 18%, respectively). We conclude the development of SPM and SHM leads to a poor survival in patients with MM and is an important survivorship challenge. Given the median survival for MM continues to improve, continued vigilance is needed to assess the risks of SPM and SHM with maintenance therapy post-auto-HSCT.
Collapse
Affiliation(s)
| | | | - Anita D’Souza
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Noel Estrada-Merly
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Lohith Gowda
- Yale Cancer Center and Yale School of Medicine, New Haven, CT
| | - Gemlyn George
- University of Colorado School of Medicine, Aurora, CO
| | - Marcos de Lima
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Shahrukh Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | - Mohamed A. Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | | | - Rahul Banerjee
- Division of Medical Oncology, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ayman Saad
- Division of Hematology, The Ohio State University, Columbus, OH
| | | | - Hira Mian
- McMaster University, Hamilton, ON, Canada
| | - Muhammad Bilal Abid
- Divisions of Hematology/Oncology & Infectious Diseases, BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI
| | | | - Lazaros J. Lekakis
- Division of Transplantation and Cellular Therapy, University of Miami Hospital and Clinics, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Sagar S. Patel
- Transplant and Cellular Therapy Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Hemant S. Murthy
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Christopher Strouse
- Division of Hematology, Oncology, and Bone & Marrow Transplantation, University of Iowa, Iowa City, IA
| | - Sherif M. Badawy
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL
| | - Samer Al Hadidi
- University of Arkansas for Medical Sciences, Little Rock, AR
| | | | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia
| | - David H. Vesole
- John Theurer Cancer Center at Hackensack Meridian School of Medicine, Hackensack, NJ
| | - Cindy H. Lee
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Attaphol Pawarode
- Adult Blood and Marrow Transplantation and Cellular Therapy, Rogel Cancer Center, Division of Hematology/Oncology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI
| | - Usama Gergis
- Department of Medical Oncology, Division of Hematological Malignancies, Thomas Jefferson University, Philadelphia, PA
| | | | - Leona A. Holmberg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Aimaz Afrough
- Myeloma, Waldenstrom's and Amyloidosis Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, GA
| | - Pashna N. Munshi
- Stem Cell Transplant and Cellular Immunotherapy Program, MedStar Georgetown University Hospital, Washington, DC
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Larry D. Anderson
- Myeloma, Waldenstrom's and Amyloidosis Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | - Baldeep Wirk
- Bone Marrow Transplant Program, Penn State Cancer Institute, Hershey, PA
| | - Gurbakhash Kaur
- Myeloma, Waldenstrom's and Amyloidosis Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | - Muzaffar H. Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Nina Shah
- Haematology Research & Development, AstraZeneca, San Francisco, CA
| | | | | |
Collapse
|
222
|
Nguyen A, Nguyen A, Dada OT, Desai PD, Ricci JC, Godbole NB, Pierre K, Lucke-Wold B. Leptomeningeal Metastasis: A Review of the Pathophysiology, Diagnostic Methodology, and Therapeutic Landscape. Curr Oncol 2023; 30:5906-5931. [PMID: 37366925 PMCID: PMC10297027 DOI: 10.3390/curroncol30060442] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
The present review aimed to establish an understanding of the pathophysiology of leptomeningeal disease as it relates to late-stage development among different cancer types. For our purposes, the focused metastatic malignancies include breast cancer, lung cancer, melanoma, primary central nervous system tumors, and hematologic cancers (lymphoma, leukemia, and multiple myeloma). Of note, our discussion was limited to cancer-specific leptomeningeal metastases secondary to the aforementioned primary cancers. LMD mechanisms secondary to non-cancerous pathologies, such as infection or inflammation of the leptomeningeal layer, were excluded from our scope of review. Furthermore, we intended to characterize general leptomeningeal disease, including the specific anatomical infiltration process/area, CSF dissemination, manifesting clinical symptoms in patients afflicted with the disease, detection mechanisms, imaging modalities, and treatment therapies (both preclinical and clinical). Of these parameters, leptomeningeal disease across different primary cancers shares several features. Pathophysiology regarding the development of CNS involvement within the mentioned cancer subtypes is similar in nature and progression of disease. Consequently, detection of leptomeningeal disease, regardless of cancer type, employs several of the same techniques. Cerebrospinal fluid analysis in combination with varied imaging (CT, MRI, and PET-CT) has been noted in the current literature as the gold standard in the diagnosis of leptomeningeal metastasis. Treatment options for the disease are both varied and currently in development, given the rarity of these cases. Our review details the differences in leptomeningeal disease as they pertain through the lens of several different cancer subtypes in an effort to highlight the current state of targeted therapy, the potential shortcomings in treatment, and the direction of preclinical and clinical treatments in the future. As there is a lack of comprehensive reviews that seek to characterize leptomeningeal metastasis from various solid and hematologic cancers altogether, the authors intended to highlight not only the overlapping mechanisms but also the distinct patterning of disease detection and progression as a means to uniquely treat each metastasis type. The scarcity of LMD cases poses a barrier to more robust evaluations of this pathology. However, as treatments for primary cancers have improved over time, so has the incidence of LMD. The increase in diagnosed cases only represents a small fraction of LMD-afflicted patients. More often than not, LMD is determined upon autopsy. The motivation behind this review stems from the increased capacity to study LMD in spite of scarcity or poor patient prognosis. In vitro analysis of leptomeningeal cancer cells has allowed researchers to approach this disease at the level of cancer subtypes and markers. We ultimately hope to facilitate the clinical translation of LMD research through our discourse.
Collapse
Affiliation(s)
- Andrew Nguyen
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Alexander Nguyen
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | - Persis D. Desai
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jacob C. Ricci
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | - Kevin Pierre
- Department of Radiology, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
223
|
Choo YS, Ooi MGM, Wang S, Hallinan JTPD. Multiple Myeloma: Case of a "Moving" Sternal Wire. Diagnostics (Basel) 2023; 13:2082. [PMID: 37370977 DOI: 10.3390/diagnostics13122082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Multiple myeloma generally occurs in older adults, with the clonal proliferation of plasma cells and accumulation of monoclonal protein resulting in a broad range of clinical manifestations and complications, including hypercalcemia, renal dysfunction, anaemia, and bone destruction (termed CRAB features). A 64-year-old man with no history of malignancy presented with an enlarging precordial lump occurring three years post-sternotomy for uneventful coronary artery bypass grafting surgery. Initial investigations showed anaemia and impaired renal function. Multimodal imaging performed for further evaluation showcases the radio-pathological features which can be encountered in haematological malignancy. Subsequent percutaneous biopsy confirmed an underlying plasma cell neoplasm, and a diagnosis of multiple myeloma was achieved. The prompt resolution of the lesions upon the initiation of treatment highlights the importance of early diagnosis and treatment.
Collapse
Affiliation(s)
- Yun Song Choo
- Department of Diagnostic Imaging, National University Hospital, Singapore 119074, Singapore
| | - Melissa Gaik-Ming Ooi
- Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore 119074, Singapore
| | - Shi Wang
- Department of Pathology, National University Hospital, Singapore 119074, Singapore
| | | |
Collapse
|
224
|
Yao Y, Ng JF, Park WD, Samur M, Morelli E, Encinas Mayoral J, Chyra Z, Xu Y, Derebail S, Epstein C, Nabet B, Chesi M, Gray NS, Young RA, Kwiatkowski N, Mitsiades C, Anderson KC, Lin CY, Munshi NC, Fulciniti M. CDK7 controls E2F- and MYC-driven proliferative and metabolic vulnerabilities in multiple myeloma. Blood 2023; 141:2841-2852. [PMID: 36877894 PMCID: PMC10315622 DOI: 10.1182/blood.2022018885] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/08/2023] Open
Abstract
Therapeutic targeting of CDK7 has proven beneficial in preclinical studies, yet the off-target effects of currently available CDK7 inhibitors make it difficult to pinpoint the exact mechanisms behind MM cell death mediated by CDK7 inhibition. Here, we show that CDK7 expression positively correlates with E2F and MYC transcriptional programs in cells from patients with multiple myeloma (MM); its selective targeting counteracts E2F activity via perturbation of the cyclin-dependent kinases/Rb axis and impairs MYC-regulated metabolic gene signatures translating into defects in glycolysis and reduced levels of lactate production in MM cells. CDK7 inhibition using the covalent small-molecule inhibitor YKL-5-124 elicits a strong therapeutic response with minimal effects on normal cells, and causes in vivo tumor regression, increasing survival in several mouse models of MM including a genetically engineered mouse model of MYC-dependent MM. Through its role as a critical cofactor and regulator of MYC and E2F activity, CDK7 is therefore a master regulator of oncogenic cellular programs supporting MM growth and survival, and a valuable therapeutic target providing rationale for development of YKL-5-124 for clinical use.
Collapse
Affiliation(s)
- Yao Yao
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Blood Disease Institute, Key Laboratory of Bone Marrow Stem Cell, Xuzhou Medical University, Xuzhou, China
| | - Jessica Fong Ng
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Woojun Daniel Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Mehmet Samur
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Eugenio Morelli
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | | - Zuzana Chyra
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Yan Xu
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Sanika Derebail
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | | - Behnam Nabet
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic, Scottsdale, AZ
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford Medical School, Stanford, CA
| | | | | | | | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | | - Nikhil C. Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Mariateresa Fulciniti
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
225
|
Keye P, Engelhardt M, Wäsch R, Böhringer D, Reinhard T. Management of Belantamab Mafodotin-Associated Keratopathy With Rigid Gas-Permeable Corneal Contact Lenses. Cornea 2023; 42:744-746. [PMID: 36728309 DOI: 10.1097/ico.0000000000003237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/08/2022] [Indexed: 02/03/2023]
Abstract
ABSTRACT Belantamab mafodotin is a relatively new drug used in the treatment of relapsed or refractory multiple myeloma. Clinical studies have shown promising responses, but ocular toxicity remains a major challenge with dose reduction or therapy discontinuation being the only available treatment option. We report a clinical case of a patient with severe keratopathy under therapy with belantamab. The use of rigid gas-permeable corneal contact lenses led to a major visual improvement and enabled therapy continuation at full dose over several months. Although this strategy may not be suitable for all patients, it provides an additional option for the treatment of ocular toxicity of this promising agent.
Collapse
Affiliation(s)
- Philip Keye
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; and
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Ralph Wäsch
- Department of Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Böhringer
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; and
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; and
| |
Collapse
|
226
|
Chakraborty R, Yi J, Rybicki L, Preussler J, Deol A, Loren A, Savani B, Jim HSL, Cerny J, Reynolds J, Whitten J, Wingard JR, McGuirk JP, Uberti J, Khera N, Stiff P, Jaglowski SM, Hashmi S, Holtan SG, Devine S, Hahn T, Whalen VL, Saber W, Wood W, Baker KS, Syrjala K, Majhail NS. Patient-Reported Outcomes in Long-Term Survivors of Autologous Hematopoietic Cell Transplantation in Multiple Myeloma. Transplant Cell Ther 2023; 29:388.e1-388.e6. [PMID: 36870388 PMCID: PMC10718487 DOI: 10.1016/j.jtct.2023.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 03/06/2023]
Abstract
The overall survival in patients with transplantation-eligible multiple myeloma has tripled over the past 2 decades, leading to a growing population of myeloma survivors. However, there is a paucity of data on health-related quality of life (HRQoL), distress, and health behaviors in long-term myeloma survivors who are in stable remission after autologous hematopoietic cell transplantation (AHCT). In this cross-sectional study using data from 2 randomized controlled trials of survivorship care plans and internet-based self-management intervention in transplantation survivors, the primary objective was to measure HRQoL (using the Short Form-12, version 2.0 [SF-12 v2]), distress (using the Cancer- and Treatment-Related Distress [CTXD] instrument), and health behaviors of myeloma survivors in stable remission after AHCT. A total of 345 patients at a median of 4 years (range, 1.4 to 11 years) post-AHCT were included. The mean SF-12 v2 Physical Component Summary (PCS) score was 45.5 ± 10.5, and the mean Mental Component Summary (MCS) score was 51.3 ± 10.1, compared with US population norms of 50 ± 10 for both (P < .001 and P = .021 for PCS and MCS comparisons, respectively). Notably, neither reached the threshold for a minimal clinically important difference. Approximately one-third of the patients had clinically significant distress based on the CTXD total score, with distress reported by 53% of the patients in the Health Burden domain, by 46% in the Uncertainty domain, by 33% in the Finances domain, by 31% in the Family Strain domain, by 21% in the Identity domain, and by 15% in the Medical Demands domain. Preventive care guidelines were adhered to by 81% of the myeloma survivors; however, adherence to exercise and diet guidelines were relatively low, at 33% and 13%, respectively. Myeloma AHCT survivors in stable remission have no clinically meaningful worsening in physical functioning compared with the general population. Survivorship programs should address ongoing distress due to health burden, uncertainty, and finances in myeloma survivors, along with evidence-based targeted interventions for modifiable health behaviors, such as nutrition and exercise.
Collapse
Affiliation(s)
- Rajshekhar Chakraborty
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York.
| | - Jean Yi
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lisa Rybicki
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | | | - Abhinav Deol
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Alison Loren
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Bipin Savani
- Division of Hematology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Jan Cerny
- Leukemia Program, University of Massachusetts Medical Center, Worcester, Massachusetts
| | - Jana Reynolds
- Baylor Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | | | - John R Wingard
- Division of Hematology and Oncology, University of Florida, Gainesville, Florida
| | - Joseph P McGuirk
- Hematologic Malignancies and Cellular Therapeutics, University of Kansas Hospital, Kansas City, Kansas
| | - Joseph Uberti
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Patrick Stiff
- Department of Hematology/Oncology, Loyola University Medical Center, Chicago, Illinois
| | - Samantha M Jaglowski
- James Cancer Hospital and Solove Research Institute, Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | - Steven Devine
- Center For International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Theresa Hahn
- Roswell Park Comprehensive Cancer Center, New York, New York
| | - Victoria L Whalen
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Wael Saber
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - William Wood
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - K Scott Baker
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Karen Syrjala
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | |
Collapse
|
227
|
Banerjee R, Williams L, Mikhael JR. Should I stay or should I go (to transplant)? Managing insufficient responses to induction in multiple myeloma. Blood Cancer J 2023; 13:89. [PMID: 37248225 DOI: 10.1038/s41408-023-00864-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023] Open
Affiliation(s)
- Rahul Banerjee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Louis Williams
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph R Mikhael
- Translational Genomics Research Institute (TGen), City of Hope, Phoenix, AZ, USA
| |
Collapse
|
228
|
Midha S, Nadeem O, Selamet U. Updates in Plasma Cell Dyscrasias and Related Monoclonal Immunoglobulin-Mediated Renal Disease. Semin Nephrol 2023; 42:151352. [PMID: 37257390 DOI: 10.1016/j.semnephrol.2023.151352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Pathogenic roles of monoclonal immunoglobulins in kidney disease have been attributed previously to malignant plasma cell and lymphoproliferative disorders such as multiple myeloma, lymphoplasmacytic lymphoma, chronic lymphocytic leukemia, or amyloid light chain amyloidosis. Improved technology, advancements in molecular diagnostics, and highly sensitive imaging techniques have established the need to redefine monoclonal gammopathies and the kidney disorders that are associated with monoclonal immunoglobulins regardless of tumor burden. This has led to the establishment of monoclonal gammopathy with renal significance (MGRS). MGRS was defined by the International Kidney and Monoclonal Gammopathy Research Group in 2012 as a clonal proliferative disorder that produces a nephrotoxic monoclonal immunoglobulin and does not meet previously defined hematological criteria for treatment of a specific malignancy. MGRS encompasses a wide array of pathologies with knowledge surrounding its incidence, prognosis, and management continuously increasing. This review examines the current evidence on the diagnosis, prognosis, pathogenesis, and therapy of plasma cell dyscrasias and related MGRS.
Collapse
Affiliation(s)
- Shonali Midha
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
| | - Omar Nadeem
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Umut Selamet
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
229
|
Costa BA, Mouhieddine TH, Ortiz RJ, Richter J. Revisiting the Role of Alkylating Agents in Multiple Myeloma: Up-to-Date Evidence and Future Perspectives. Crit Rev Oncol Hematol 2023; 187:104040. [PMID: 37244325 DOI: 10.1016/j.critrevonc.2023.104040] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023] Open
Abstract
From the 1960s to the early 2000s, alkylating agents (e.g., melphalan, cyclophosphamide, and bendamustine) remained a key component of standard therapy for newly-diagnosed or relapsed/refractory multiple myeloma (MM). Later on, their associated toxicities (including second primary malignancies) and the unprecedented efficacy of novel therapies have led clinicians to increasingly consider alkylator-free approaches. Meanwhile, new alkylating agents (e.g., melflufen) and new applications of old alkylators (e.g., lymphodepletion before chimeric antigen receptor T-cell [CAR-T] therapy) have emerged in recent years. Given the expanding use of antigen-directed modalities (e.g., monoclonal antibodies, bispecific antibodies, and CAR-T therapy), this review explores the current and future role of alkylating agents in different treatment settings (e.g., induction, consolidation, stem cell mobilization, pre-transplant conditioning, salvage, bridging, and lymphodepleting chemotherapy) to ellucidate the role of alkylator-based regimens in modern-day MM management.
Collapse
Affiliation(s)
- Bruno Almeida Costa
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tarek H Mouhieddine
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo J Ortiz
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
230
|
Lin CM, Chang LC, Shau WY, Chen CL, Yao CY, Tien FM. Treatment benefit of upfront autologous stem cell transplantation for newly diagnosed multiple myeloma: a systematic review and meta-analysis. BMC Cancer 2023; 23:446. [PMID: 37193978 DOI: 10.1186/s12885-023-10907-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/30/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Upfront high-dose therapy (HDT) followed by autologous stem cell transplantation (ASCT) remains a profitable strategy for newly diagnosed multiple myeloma (MM) patients in the context of novel agents. However, current knowledge demonstrates a discrepancy between progression-free survival (PFS) and overall survival (OS) benefit with HDT/ASCT. METHODS We conducted a systematic review and meta-analysis that included both randomized controlled trials (RCTs) and observational studies evaluating the benefit of upfront HDT/ASCT published during 2012 to 2023. Further sensitivity analysis and meta-regression were also performed. RESULTS Among the 22 enrolled studies, 7 RCTs and 9 observational studies had a low or moderate risk of bias, while the remaining 6 observational studies had a serious risk of bias. HDT/ASCT revealed advantages in complete response (CR) with an odds ratio (OR) of 1.24 and 95% confidence interval (CI) 1.02 ~ 1.51, PFS with a hazard ratio (HR) of 0.53 (95% CI 0.46 ~ 0.62), and OS with an HR of 0.58 (95% CI 0.50 ~ 0.69). Sensitivity analysis excluding the studies with serious risk of bias and trim-and-fill imputation fundamentally confirmed these findings. Older age, increased percentage of patients with International Staging System (ISS) stage III or high-risk genetic features, decreased proteasome inhibitor (PI) or combined PI/ immunomodulatory drugs (IMiD) utilization, and decreased follow-up duration or percentage of males were significantly related to a greater survival advantage with HDT/ASCT. CONCLUSIONS Upfront ASCT remains a beneficial treatment for newly diagnosed MM patients in the period of novel agents. Its advantage is especially acute in high-risk MM populations, such as elderly individuals, males, those with ISS stage III or high-risk genetic features, but is attenuated with PI or combined PI/IMiD utilization, contributing to divergent survival outcomes.
Collapse
Affiliation(s)
- Chi-Maw Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, Taiwan
| | - Lih-Chyun Chang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Hospital Medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Yi Shau
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Ling Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Yuan Yao
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100225, Taiwan
| | - Feng-Ming Tien
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Zhongshan S. Rd., Zhongzheng Dist., Taipei City, 100225, Taiwan.
| |
Collapse
|
231
|
Gillich C, Akhoundova D, Hayoz M, Aebi Y, Largiadèr CR, Seipel K, Daskalakis M, Bacher U, Pabst T. Efficacy and Safety of High-Dose Chemotherapy with Treosulfan and Melphalan in Multiple Myeloma. Cancers (Basel) 2023; 15:2699. [PMID: 37345036 DOI: 10.3390/cancers15102699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
(1) Background: Upfront treatment consolidation with high-dose chemotherapy (HDCT) and autologous stem cell transplantation (ASCT) has relevantly contributed to achieving durable remissions following induction treatment in multiple myeloma (MM) patients. The optimization of HDCT regimens can, therefore, essentially contribute to improving the depth and duration of tumor remissions. To date, melphalan at 200 mg/m2 is the standard HDCT regimen for fit MM patients. In our previous work, we showed promising efficacy and safety results for treosulfan (14 g/m2) and melphalan (200 mg/m2) (TreoMel) in acute myeloid leukemia (AML) patients receiving ASCT. Based on these data, TreoMel became the standard of care for fit MM patients at our institution. (2) Methods: We identified 115 consecutive MM patients who underwent consolidation with TreoMel between 01/2020 and 08/2022 at the University Hospital of Bern. We analyzed the safety and efficacy data, as well as the treosulfan pharmacokinetics, correlating them with tumor responses. (3) Results: A complete response (CR) rate of 84% was achieved, which is comparable to the CR rate reported for the quadruplet combination. The median PFS was 30 months (95% CI: 20.4-not reached), and the 31-month OS rate was 83%. The median area under the curve (AUC) for treosulfan was 952.5 mg*h/L (range: 527.4-1781.4), and the median peak level was 332.3 mg/L (range: 168-554). The treosulfan pharmacokinetics showed no significant correlation with MM responses after HDCT and ASCT. However, female patients had a significantly higher AUC (p = 0.007) and peak value (p = 0.001), and the higher values were associated with longer hospitalizations. (4) Conclusions: Treatment consolidation with TreoMel HDCT demonstrated a promising efficacy and safety profile in our cohort of MM patients and deserves further investigation in prospective studies.
Collapse
Affiliation(s)
- Cédric Gillich
- Department of Medical Oncology, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Dilara Akhoundova
- Department of Medical Oncology, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Michael Hayoz
- Department of Clinical Chemistry and Center for Laboratory Medicine, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
- University Institute of Clinical Chemistry, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Yolanda Aebi
- Department of Clinical Chemistry and Center for Laboratory Medicine, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
- University Institute of Clinical Chemistry, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Carlo R Largiadèr
- Department of Clinical Chemistry and Center for Laboratory Medicine, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
- University Institute of Clinical Chemistry, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Katja Seipel
- Department of Medical Oncology, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Michael Daskalakis
- Department of Hematology, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Ulrike Bacher
- Department of Hematology, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
232
|
Drummond PLM, Santos RMMD, Reis AMM, Malta JS, Silveira LP, Costa IHFD, Menezes de Pádua CA. Real-world effectiveness and safety of multiple myeloma treatments based on thalidomide and bortezomib: A retrospective cohort study from 2009 to 2020 in a Brazilian metropolis. Cancer Epidemiol 2023; 85:102377. [PMID: 37163919 DOI: 10.1016/j.canep.2023.102377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/27/2023] [Accepted: 04/29/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Multiple myeloma (MM) is an incurable cancer of plasma cells; the survival of which has improved over the years with the emergence of new treatments. In Brazil, the availability of treatment-regimens is different from developed countries. Real-world evidence with Brazilian patients is lacking. OBJECTIVES Our aim was to evaluate the effectiveness and the safety of MM treatments in a Brazilian metropolis. METHODS This was a retrospective cohort study with MM patients, beginning MM treatment from 2009 to 2020 (i.e., before bortezomib became available in public health services). Patients' medical records were revised to obtain clinical variables. The primary outcomes were Overall Survival (OS) and Progression Free Survival (PFS, measured as time to next treatment), and the secondary outcomes were Adverse Events (AE). Kaplan-Meier curves were obtained and the Cox proportional hazards model was performed for univariate and multivariate analyses. The incidence of AE was estimated and the chi-squared test was performed to evaluate the association between AE and MM regimens. RESULTS In total, 278 patients participated in the study with median age of 64 years; 50.4 % were females, 55.8 % attended a private clinic, 34.9 % received autologous stem cell transplantation (ASCT) and 32.4 % were on polypharmacy. Most patients from public services used thalidomide-based regimens (40.3 %) and at private clinics used bortezomib-based regimens (38.1 %) as first-line treatment. Patients had a median OS of 99 months. Patients had median PFS of 28 months in first-line treatment, which was significantly different for age (p = 0.0055), polypharmacy (p = 0.0094) and ASCT (p < 0.0001). PFS was independently associated to polypharmacy and ASCT. The incidence of peripheral neuropathy (39.6 %) was high. In contrast, the incidence of severe AE was low. We found significant difference between first-line T + B-based regimens and leukopenia (p = 0.012). CONCLUSION Our study showed that patients on polypharmacy and who did not receive ASCT had worse PFS. Similar to other Latin countries, most patients used thalidomide- and bortezomib-based regimens as first-line treatments having similar OS and PFS. Treatments were considered relatively safe, especially regarding serious AE.
Collapse
Affiliation(s)
- P L M Drummond
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Brazil; Ezequiel Dias Foundation, Brazil
| | | | | | | | - Lívia Pena Silveira
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Brazil; Hospital das Clínicas, Universidade Federal de Minas Gerais, Brazil
| | | | | |
Collapse
|
233
|
Mohyuddin GR, Goodman AM. Ineligible for transplant, but eligible for intensive quadruplet therapy—The value of ‘add-on’ trials for vulnerable patient populations. Eur J Cancer 2023; 185:164-166. [PMID: 36996626 DOI: 10.1016/j.ejca.2023.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/30/2023]
Affiliation(s)
- Ghulam Rehman Mohyuddin
- Division of Hematology and Hematological Malignancies, University of Utah, Salt Lake City, Utah, USA; Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California, USA.
| | - Aaron M Goodman
- Division of Hematology and Hematological Malignancies, University of Utah, Salt Lake City, Utah, USA; Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
234
|
Rejeski K, Greco R, Onida F, Sánchez-Ortega I, Bonini C, Sureda A, Gribben JG, Yakoub-Agha I, Subklewe M. An International Survey on Grading, Diagnosis, and Management of Immune Effector Cell-Associated Hematotoxicity (ICAHT) Following CAR T-cell Therapy on Behalf of the EBMT and EHA. Hemasphere 2023; 7:e889. [PMID: 37125259 PMCID: PMC10145722 DOI: 10.1097/hs9.0000000000000889] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
Hematological toxicity represents the most common grade ≥3 toxicity after chimeric antigen receptor (CAR) T-cell therapy. However, its underlying pathophysiology is incompletely understood and its grading and management remains ill-defined. To inform the forthcoming European Hematology Association/European Society for Blood and Marrow Transplantation (EHA/EBMT) guidelines on the management of "immune effector cell-associated hematotoxicity" (ICAHT), we undertook a survey of experienced clinicians using an online survey focusing on (1) grading, (2) risk-stratification and diagnostic work-up, (3) short-term, and (4) long-term management of ICAHT. There were 81 survey respondents across 18 countries. A high degree of variability was noted for cytopenia grading in regards to depth, duration, and time from CAR-T infusion. The majority of experts favored pre-CAR-T bone marrow studies, especially in case of a high-risk profile. Most respondents felt that the work-up for patients with severe hematotoxicity should rule-out viral infections (96%), substrate deficiency (80%), or coincident sHLH/MAS (serum ferritin, 92%), and should include bone marrow aspiration (86%) and/or biopsy (61%). Clinicians were divided as to whether the occurrence of coincident immunotoxicity should influence the decision to apply G-CSF, and when to initiate G-CSF support. In case of prolonged thrombocytopenia, most survey participants favored thrombopoietin agonists (86%). Conversely, autologous hematopoietic cell boosts represented the preferred choice for neutropenia (63%), although they were frequently not available and no consensus was reached regarding the optimal trigger point. These findings underline the current heterogeneity of practice patterns regarding ICAHT and invite the development of consensus guidelines, which may harmonize grading, establish standard operating procedures for diagnosis, and set management guidelines.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Onida
- Hematology and BMT Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Italy
| | - Isabel Sánchez-Ortega
- European Society for Blood and Marrow Transplantation (EBMT) Executive Office, Barcelona, Spain
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Anna Sureda
- Institut Català d’Oncologia-Hospital Duran i Reynals, Barcelona, Spain
| | - John G. Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University London, United Kingdom
| | | | - Marion Subklewe
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Germany
| |
Collapse
|
235
|
Suzuki K, Yano S. Treatment Strategy for Ultra-High-Risk Multiple Myelomas with Chromosomal Aberrations Considering Minimal Residual Disease Status and Bone Marrow Microenvironment. Cancers (Basel) 2023; 15:cancers15092418. [PMID: 37173885 PMCID: PMC10177433 DOI: 10.3390/cancers15092418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the development of anti-myeloma therapeutics, such as proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies, and autologous stem cell transplantation (ASCT), multiple myeloma remains incurable. A trial treatment combining four drugs-daratumumab, carfilzomib, lenalidomide, and dexamethasone-followed by ASCT frequently results in minimal residual disease (MRD) negativity and prevents progressive disease in patients with standard- and high-risk cytogenetics; however, it is insufficient to overcome the poor outcomes in patients with ultra-high-risk chromosomal aberration (UHRCA). In fact, MRD status in autografts can predict clinical outcomes after ASCT. Therefore, the current treatment strategy might be insufficient to overcome the negative impact of UHRCA in patients with MRD positivity after the four-drug induction therapy. High-risk myeloma cells lead to poor clinical outcomes not only by aggressive myeloma behavior but also via the generation of a poor bone marrow microenvironment. Meanwhile, the immune microenvironment effectively suppresses myeloma cells with a low frequency of high-risk cytogenetic abnormalities in early-stage myeloma compared to late-stage myeloma. Therefore, early intervention might be key to improving clinical outcomes in myeloma patients. The purpose of this review is to improve clinical outcomes in patients with UHRCA by considering MRD assessment results and improvement of the microenvironment.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
236
|
Kawamura K. Maintenance therapy after allogeneic hematopoietic stem cell transplantation for patients with multiple myeloma. Int J Hematol 2023:10.1007/s12185-023-03602-1. [PMID: 37060508 DOI: 10.1007/s12185-023-03602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/16/2023]
Abstract
In the last two decades, proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and monoclonal antibodies have greatly improved the overall survival of patients with multiple myeloma. However, multiple myeloma remains incurable, and high-risk patients have poor long-term survival. Although allogeneic hematopoietic stem cell transplantation (allo-HCT) is not considered standard therapy because of relatively high transplant-related mortality and relapse rates, the graft-versus-myeloma (GVM) effect makes it a potentially curative therapy. Therefore, allo-HCT remains a treatment option for younger patients and those with high-risk myeloma. Maintenance therapy with novel agents has recently been attempted to reduce relapse in patients undergoing allo-HCT, but its effectiveness remains unclear. This review focuses on the role of maintenance therapy after allo-HCT in patients with myeloma. Maintenance therapy using IMiDs and/or PIs after allo-HCT may be effective in reducing relapse or improving response because it may prevent early progression before achievement of the GVM effect or enhance the GVM effect. However, care must be taken to avoid complications, such as graft-versus-host disease. Further studies are necessary to determine the optimal maintenance drugs, drug combinations, dosing, start timing, and number of cycles.
Collapse
Affiliation(s)
- Koji Kawamura
- Division of Clinical Laboratory Medicine, Department of Multidisciplinary Internal Medicine, Tottori University, 86, Nishi-Cho, Yonago-City, Tottori, 683-8503, Japan.
| |
Collapse
|
237
|
Yin Z, Lv Y, Deng L, Li G, Ou R, Chen L, Zhu Y, Zhong Q, Liu Z, Huang J, Wu H, Zhang Q, Fei J, Liu S. Targeting ABCB6 with nitidine chloride inhibits PI3K/AKT signaling pathway to promote ferroptosis in multiple myeloma. Free Radic Biol Med 2023; 203:86-101. [PMID: 37044150 DOI: 10.1016/j.freeradbiomed.2023.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/19/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Since multiple myeloma (MM) remains a cureless malignancy of plasma cells to date, it becomes imperative to develop novel drugs and therapeutic targets for MM. We screened a small molecule library comprising 3633 natural product drugs, which demonstrated that Nitidine Chloride (NC), an extract from traditional Chinese medicine Zanthoxylum nitidum. We used Surface Plasmon Resonance-High Performance Liquid Chromatography-Protein Mass Spectrometry (SPR-HPLC-MS), Cellular Thermal Shift Assay (CETSA), molecular docking, and SPR assay to identify the potential targets of NC, in which ABCB6 was the unique target of NC. The effects of ABCB6 on cellular proliferation and drug resistance were determined by CCK8, western blot, flow cytometry, site-mutation cells, transmission electron microscopy, immunohistochemistry staining and xenograft model in vitro and in vivo. NC induced MM cell death by promoting ferroptosis. ABCB6 is the direct target of NC. ABCB6 expression was increased in MM samples compared to normal controls, which was significantly associated with MM relapse and poor outcomes. VGSK was the inferred binding epitope of NC on the ABCB6 protein. In the ABCB6-mutated MM cells, NC did not display cancer resistance, implying the vital role of ABCB6 in NC's bioactivity. Moreover, the silencing of ABCB6 significantly inhibited MM cell growth. Mechanistically, the direct binding of NC to ABCB6 suppressed PI3K/AKT signaling pathway to promote ferroptosis. In conclusion, ABCB6 can be a potential therapeutic target and prognostic biomarker in MM, while NC can be considered a novel drug for MM treatment.
Collapse
Affiliation(s)
- Zhao Yin
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Yiwen Lv
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Guangchao Li
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Ruiming Ou
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Lizhi Chen
- Department of Science and Education, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Yangmin Zhu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Qi Zhong
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Zhi Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Jing Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Hong Wu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Jia Fei
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou, 510632, China
| | - Shuang Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China.
| |
Collapse
|
238
|
Morè S, Corvatta L, Manieri VM, Olivieri A, Offidani M. Current Main Topics in Multiple Myeloma. Cancers (Basel) 2023; 15:2203. [PMID: 37190132 PMCID: PMC10136770 DOI: 10.3390/cancers15082203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Multiple Myeloma (MM) remains a difficult to treat disease mainly due to its biological heterogeneity, of which we are more and more knowledgeable thanks to the development of increasingly sensitive molecular methods that allow us to build better prognostication models. The biological diversity translates into a wide range of clinical outcomes from long-lasting remission in some patients to very early relapse in others. In NDMM transplant eligible (TE) patients, the incorporation of mAb as daratumumab in the induction regimens, followed by autologous stem cell transplantation (ASCT) and consolidation/maintenance therapy, has led to a significant improvement of PFS and OS.; however, this outcome remains poor in ultra-high risk MM or in those who did not achieve a minimal residual disease (MRD) negativity. Several trials are exploring cytogenetic risk-adapted and MRD-driven therapies in these patients. Similarly, quadruplets-containing daratumumab, particularly when administered as continuous therapies, have improved outcome of patients not eligible for autologous transplant (NTE). Patients who become refractory to conventional therapies have noticeably poor outcomes, making their treatment a difficult challenge in need of novel strategies. In this review, we will focus on the main points regarding risk stratification, treatment and monitoring of MM, highlighting the most recent evidence that could modify the management of this still incurable disease.
Collapse
Affiliation(s)
- Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| | - Laura Corvatta
- Unità Operativa Complessa di Medicina, Ospedale Profili, 60044 Fabriano, Italy
| | | | - Attilio Olivieri
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| | - Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| |
Collapse
|
239
|
Samur MK, Roncador M, Aktas Samur A, Fulciniti M, Bazarbachi AH, Szalat R, Shammas MA, Sperling AS, Richardson PG, Magrangeas F, Minvielle S, Perrot A, Corre J, Moreau P, Thakurta A, Parmigiani G, Anderson KC, Avet-Loiseau H, Munshi NC. High-dose melphalan treatment significantly increases mutational burden at relapse in multiple myeloma. Blood 2023; 141:1724-1736. [PMID: 36603186 PMCID: PMC10273091 DOI: 10.1182/blood.2022017094] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
High-dose melphalan (HDM) improves progression-free survival in multiple myeloma (MM), yet melphalan is a DNA-damaging alkylating agent; therefore, we assessed its mutational effect on surviving myeloma cells by analyzing paired MM samples collected at diagnosis and relapse in the IFM 2009 study. We performed deep whole-genome sequencing on samples from 68 patients, 43 of whom were treated with RVD (lenalidomide, bortezomib, and dexamethasone) and 25 with RVD + HDM. Although the number of mutations was similar at diagnosis in both groups (7137 vs 7230; P = .67), the HDM group had significantly more mutations at relapse (9242 vs 13 383, P = .005). No change in the frequency of copy number alterations or structural variants was observed. The newly acquired mutations were typically associated with DNA damage and double-stranded breaks and were predominantly on the transcribed strand. A machine learning model, using this unique pattern, predicted patients who would receive HDM with high sensitivity, specificity, and positive prediction value. Clonal evolution analysis showed that all patients treated with HDM had clonal selection, whereas a static progression was observed with RVD. A significantly higher percentage of mutations were subclonal in the HDM cohort. Intriguingly, patients treated with HDM who achieved complete remission (CR) had significantly more mutations at relapse yet had similar survival rates as those treated with RVD who achieved CR. This similarity could have been due to HDM relapse samples having significantly more neoantigens. Overall, our study identifies increased genomic changes associated with HDM and provides rationale to further understand clonal complexity.
Collapse
Affiliation(s)
- Mehmet Kemal Samur
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | | | - Anil Aktas Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Abdul Hamid Bazarbachi
- Department of Internal Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, New York, NY
| | - Raphael Szalat
- Department of Hematology and Medical Oncology, Boston University Medical Center, Boston, MA
| | - Masood A. Shammas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Adam S. Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Paul G. Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Florence Magrangeas
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | - Stephane Minvielle
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | - Aurore Perrot
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Jill Corre
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Philippe Moreau
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | | | - Giovanni Parmigiani
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | - Kenneth C. Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Hervé Avet-Loiseau
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Nikhil C. Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
240
|
Cole K, Al-Kadhimi Z, Talmadge JE. Highlights into historical and current immune interventions for cancer. Int Immunopharmacol 2023; 117:109882. [PMID: 36848790 PMCID: PMC10355273 DOI: 10.1016/j.intimp.2023.109882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Immunotherapy is an additional pillar when combined with traditional standards of care such as chemotherapy, radiotherapy, and surgery for cancer patients. It has revolutionized cancer treatment and rejuvenated the field of tumor immunology. Several types of immunotherapies, including adoptive cellular therapy (ACT) and checkpoint inhibitors (CPIs), can induce durable clinical responses. However, their efficacies vary, and only subsets of cancer patients benefit from their use. In this review, we address three goals: to provide insight into the history of these approaches, broaden our understanding of immune interventions, and discuss current and future approaches. We highlight how cancer immunotherapy has evolved and discuss how personalization of immune intervention may address present limitations. Cancer immunotherapy is considered a recent medical achievement and in 2013 was selected as the "Breakthrough of the Year" by Science. While the breadth of immunotherapeutics has been rapidly expanding, to include the use of chimeric antigen receptor (CAR) T-cell therapy and immune checkpoint inhibitor (ICI) therapy, immunotherapy dates back over 3000 years. The expansive history of immunotherapy, and related observations, have resulted in several approved immune therapeutics beyond the recent emphasis on CAR-T and ICI therapies. In addition to other classical forms of immune intervention, including human papillomavirus (HPV), hepatitis B, and the Mycobacterium bovis Bacillus Calmette-Guérin (BCG) tuberculosis vaccines, immunotherapies have had a broad and durable impact on cancer therapy and prevention. One classic example of immunotherapy was identified in 1976 with the use of intravesical administration of BCG in patients with bladder cancer; resulting in a 70 % eradication rate and is now standard of care. However, a greater impact from the use of immunotherapy is documented by the prevention of HPV infections that are responsible for 98 % of cervical cancer cases. In 2020, the World Health Organization (WHO) estimated that 341,831 women died from cervical cancer [1]. However, administration of a single dose of a bivalent HPV vaccine was shown to be 97.5 % effective in preventing HPV infections. These vaccines not only prevent cervical squamous cell carcinoma and adenocarcinoma, but also oropharyngeal, anal, vulvar, vaginal, and penile squamous cell carcinomas. The breadth, response and durability of these vaccines can be contrasted with CAR-T-cell therapies, which have significant barriers to their widespread use including logistics, manufacturing limitations, toxicity concerns, financial burden and lasting remissions observed in only 30 to 40 % of responding patients. Another, recent immunotherapy focus are ICIs. ICIs are a class of antibodies that can increase the immune responses against cancer cells in patients. However, ICIs are only effective against tumors with a high mutational burden and are associated with a broad spectrum of toxicities requiring interruption of administration and/or administration corticosteroids; both of which limit immune therapy. In summary, immune therapeutics have a broad impact worldwide, utilizing numerous mechanisms of action and when considered in their totality are more effective against a broader range of tumors than initially considered. These new cancer interventions have tremendous potential notability when multiple mechanisms of immune intervention are combined as well as with standard of care modalities.
Collapse
Affiliation(s)
- Kathryn Cole
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zaid Al-Kadhimi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
241
|
Engelhardt M, Brioli A, von Lilienfeld-Toal M. [Differences due to socio-economic status, genetic background and sex in cancer and precision medicine - An intersectional approach to close the care gap for marginalized groups]. Dtsch Med Wochenschr 2023; 148:528-538. [PMID: 37094588 DOI: 10.1055/a-1892-4833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Malignant tumor diseases constitute the 2nd most common cause of death and due to our extended life expectancy cancer per se has substantially increased, being highly prevalent after cardiovascular diseases. Evidence also generated from the COVID-19 pandemic, that defined gender differences exist in symptom and disease courses, and have advocated the need to assess gender, ethnic/racial and minority differences in cancer care and treatment more meticulously. It is becoming increasingly evident that in novel cancer care/precision oncology, representation of minorities, elderly and frail patients in clinical trials remains largely unbalanced, thus distribution of cancer success is iniquitous. This article focusses on these aspects and suggests solutions, how this can be improved.
Collapse
|
242
|
Hashmi H, Atrash S, Jain J, Khasawneh G, Mohan M, Mahmoudjafari Z, Cui W, McGuirk J, Shune L, Ahmed N, Abdallah AO. Daratumumab, pomalidomide, and dexamethasone (DPd) followed by high dose chemotherapy-Autologous Stem Cell Transplantation leads to superior outcomes when compared to DPd-alone for patients with Relapsed Refractory Multiple Myeloma. Transplant Cell Ther 2023; 29:262.e1-262.e6. [PMID: 36682468 DOI: 10.1016/j.jtct.2023.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/26/2022] [Accepted: 01/15/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND OBJECTIVES While the role of autologous stem cell transplant (ASCT) in the first line therapy for newly diagnosed multiple myeloma is well established, efficacy of ASCT for patients with relapsed refractory multiple myeloma (RRMM) in the era of novel therapeutic agents remains unknown. In this single center retrospective analysis, we evaluated and compared the efficacy and safety outcomes of patients with RRMM treated with daratumumab pomalidomide dexamethasone (DPd) alone versus (vs) DPd followed by ASCT. METHODS A total of 83 patients with RRMM who were treated with and achieved at least partial response (PR) with DPd were evaluated by electronic medical records. All patients who responded to DPd and were deemed eligible for ASCT proceeded with high dose melphalan followed by autologous stem cell infusion (DPd + ASCT group). Remaining patients continued DPd until disease progression or intolerable toxicities (DPd-alone group). Responses were evaluated using the International Myeloma Working Group response criteria and toxicities were graded using National Cancer Institute Common Terminology Criteria for Adverse Events. Patient and disease characteristics, as well as efficacy and safety outcomes were summarized using descriptive statistics. Kaplan-Meier analyses were used to estimate progression-free survival (PFS) and overall survival (OS). RESULTS A total of 21/83 (25%) patients with RRMM who achieved at least PR to DPd underwent ASCT (DPd + ASCT group) while the remaining 62/83 (75%) continued DPd without ASCT (DPd-alone group). For the entire patient population, median age was 66 years (42-81), 49 (59%) patients were male, 54 (65%) patients had IgG isotype, 21 (25%) patients had R-ISS stage III disease, 51 (61%) patients had high-risk cytogenetics, and 17 (20%) patients had extramedullary disease. Patient age, disease stage, cytogenetic risk profile were well balanced between two groups. A stringent complete response was seen in 10 (16%) and 12 (57%) patients in the DPd-alone and DPd + AST groups, respectively. Median PFS was 17.5 months in the DPd-alone vs 42.2 months (p=0.006) in the DPd + ASCT group. Median OS was 38.1 months in the DPd-alone group vs not reached in the DPD + ASCT group (p=0.009). The most common grade 3 or 4 treatment-related adverse events (TRAE) were myelosuppression and gastrointestinal toxicities, more commonly seen in the DPd + ASCT group. No treatment-related mortalities were observed in either group. CONCLUSION Patients with RRMM who responded to DPd and underwent HDT-ASCT demonstrated superior depth and duration of remission compared to those who received DPd-alone. Although DPd followed by ASCT is associated with more cytopenias and gastrointestinal toxicities, this treatment appears to be overall safe for patients with RRMM.
Collapse
Affiliation(s)
- Hamza Hashmi
- Division of Hematology/Oncology, Medical University of South Carolina, Charleston, SC, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US.
| | - Shebli Atrash
- Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| | - Jayanshu Jain
- University of Kansas Medical Center, Westwood, KS, US
| | - Ghena Khasawneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Meera Mohan
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| | - Zahra Mahmoudjafari
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| | - Wei Cui
- University of Kansas Medical Center, Westwood, KS, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| | - Joseph McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, US
| | - Leyla Shune
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| | - Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| | - Al-Ola Abdallah
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, US; US Myeloma Research Innovations Research Collaborative (USMIRC), Westwood, KS, US
| |
Collapse
|
243
|
Raje N, Mateos MV, Iida S, Reece D. Clinical evidence for immune-based strategies in early-line multiple myeloma: current challenges in decision-making for subsequent therapy. Blood Cancer J 2023; 13:41. [PMID: 36944635 PMCID: PMC10030780 DOI: 10.1038/s41408-023-00804-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/07/2023] [Accepted: 02/23/2023] [Indexed: 03/23/2023] Open
Abstract
Almost all patients with multiple myeloma (MM) will eventually develop disease that has relapsed with or become refractory to available treatments and will require additional therapy. However, data are still lacking on how best to sequence regimens in the relapsed/refractory (RR) setting after the failure of early-line lenalidomide, bortezomib, and/or daratumumab, the most commonly used agents in clinical practice today. With the treatment landscape rapidly changing in response to emerging clinical trial data and approvals of several new drugs and additional combinations, it is critically important to focus on patients with RRMM. Variability in patient baseline characteristics, such as the number of prior lines of treatment, refractoriness to prior treatments, prior stem cell transplant, and timing and dosing of prior lenalidomide, makes it difficult to select the best options for patients with RRMM for whom first-line treatments have failed. The aim of this review is to provide both an overview of current therapies and future directions within the RRMM treatment landscape, and a framework for clinicians to choose the most promising next treatment option.
Collapse
Affiliation(s)
- Noopur Raje
- Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA.
| | - María-Victoria Mateos
- University Hospital of Salamanca/IBSAL/Cancer Research Center-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Donna Reece
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
244
|
Burgos L, Tamariz-Amador LE, Puig N, Cedena MT, Guerrero C, Jelínek T, Johnson S, Milani P, Cordon L, Perez JJ, Lasa M, Termini R, Oriol A, Hernandez MT, Palomera L, Martinez-Martinez R, de la Rubia J, de Arriba F, Rios R, Gonzalez ME, Gironella M, Cabañas V, Casanova M, Krsnik I, Perez-Montaña A, González-Calle V, Rodriguez-Otero P, Maisnar V, Hajek R, Van Rhee F, Jimenez-Zepeda V, Palladini G, Merlini G, Orfao A, de la Cruz J, Martinez-Lopez J, Lahuerta JJ, Rosiñol L, Blade J, Mateos MV, San-Miguel JF, Paiva B. Definition and Clinical Significance of the Monoclonal Gammopathy of Undetermined Significance-Like Phenotype in Patients With Monoclonal Gammopathies. J Clin Oncol 2023; 41:3019-3031. [PMID: 36930848 DOI: 10.1200/jco.22.01916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
PURPOSE The existence of patients with multiple myeloma (MM) and light-chain (AL) amyloidosis who present with a monoclonal gammopathy of undetermined significance (MGUS)-like phenotype has been hypothesized, but methods to identify this subgroup are not standardized and its clinical significance is not properly validated. PATIENTS AND METHODS An algorithm to identify patients having MGUS-like phenotype was developed on the basis of the percentages of total bone marrow (BM) plasma cells (PC) and of clonal PC within the BM PC compartment, determined at diagnosis using flow cytometry in 548 patients with MGUS and 2,011 patients with active MM. The clinical significance of the algorithm was tested and validated in 488 patients with smoldering MM, 3,870 patients with active MM and 211 patients with AL amyloidosis. RESULTS Patients with smoldering MM with MGUS-like phenotype showed significantly lower rates of disease progression (4.5% and 0% at 2 years in two independent series). There were no statistically significant differences in time to progression between treatment versus observation in these patients. In active newly diagnosed MM, MGUS-like phenotype retained independent prognostic value in multivariate analyses of progression-free survival (PFS; hazard ratio [HR], 0.49; P = .001) and overall survival (OS; HR, 0.56; P = .039), together with International Staging System, lactate dehydrogenase, cytogenetic risk, transplant eligibility, and complete remission status. Transplant-eligible patients with active MM with MGUS-like phenotype showed PFS and OS rates at 5 years of 79% and 96%, respectively. In this subgroup, there were no differences in PFS and OS according to complete remission and measurable residual disease status. Application of the algorithm in two independent series of patients with AL predicted for different survival. CONCLUSION We developed an open-access algorithm for the identification of MGUS-like patients with distinct clinical outcomes. This phenotypic classification could become part of the diagnostic workup of MM and AL amyloidosis.
Collapse
Affiliation(s)
- Leire Burgos
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Luis-Esteban Tamariz-Amador
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Noemi Puig
- Hospital Universitario de Salamanca (HUSAL), IBSAL, IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Maria-Teresa Cedena
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC, Instituto de Investigación IMAS12, Madrid, Spain
| | - Camila Guerrero
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Tomas Jelínek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Sarah Johnson
- Myeloma Center/Division of Hematology Oncology/Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Paolo Milani
- Department of Molecular Medicine, University of Pavia and Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Jose J Perez
- Hospital Universitario de Salamanca (HUSAL), IBSAL, IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Marta Lasa
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Rosalinda Termini
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Albert Oriol
- Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Barcelona, Spain
| | | | - Luis Palomera
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | | | | | - Felipe de Arriba
- Hospital Morales Meseguer, IMIB-Arrixaca, Universidad de Murcia, Murcia, Spain
| | - Rafael Rios
- Hospital Universitario Puerta de Hierro, Hospital, Madrid, Spain
| | | | - Mercedes Gironella
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Valentin Cabañas
- Hospital Clínico Universitario Virgen de la Arrixaca. IMIB-Arrixaca. University of Murcia, Murcia, Spain
| | - Maria Casanova
- Hematology Department, Hospital Costa del Sol Marbella, Marbella, Spain
| | - Isabel Krsnik
- Hospital Universitario Puerta de Hierro, Hospital, Madrid, Spain
| | | | - Verónica González-Calle
- Hospital Universitario de Salamanca (HUSAL), IBSAL, IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Paula Rodriguez-Otero
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Vladimir Maisnar
- 4th Department of Medicine-Haematology, Charles University Hospital, Hradec Králové, Czech Republic
| | - Roman Hajek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Fritz Van Rhee
- Myeloma Center/Division of Hematology Oncology/Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Victor Jimenez-Zepeda
- Tom Baker Cancer Center, Department of Hematology, University of Calgary, Calgary, AB, Canada
| | - Giovanni Palladini
- Department of Molecular Medicine, University of Pavia and Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giampaolo Merlini
- Department of Molecular Medicine, University of Pavia and Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alberto Orfao
- Hospital Universitario de Salamanca (HUSAL), IBSAL, Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service, University of Salamanca, CIBERONC, Salamanca, Spain
| | - Javier de la Cruz
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC, Instituto de Investigación IMAS12, Madrid, Spain
| | - Joaquin Martinez-Lopez
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC, Instituto de Investigación IMAS12, Madrid, Spain
| | - Juan-Jose Lahuerta
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC, Instituto de Investigación IMAS12, Madrid, Spain
| | - Laura Rosiñol
- Amyloidosis and Myeloma Unit. Department of Hematology. Hospital Clínic de Barcelona. IDIBAPS., Barcelona, Spain
| | - Joan Blade
- Amyloidosis and Myeloma Unit. Department of Hematology. Hospital Clínic de Barcelona. IDIBAPS., Barcelona, Spain
| | - Maria-Victoria Mateos
- Hospital Universitario de Salamanca (HUSAL), IBSAL, IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Jesus F San-Miguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | | |
Collapse
|
245
|
Novel Agents as Main Drivers for Continued Improvement in Survival in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15051558. [PMID: 36900349 PMCID: PMC10000382 DOI: 10.3390/cancers15051558] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
(1) Background: New therapeutic strategies have improved the prognosis of multiple myeloma (MM), changing the accepted view of this disease from being incurable to treatable. (2) Methods: We studied 1001 patients with MM between 1980 and 2020, grouping patients into ten-year periods by diagnosis 1980-1990, 1991-2000, 2001-2010 and 2011-2020. (3) Results: After 65.1 months of follow-up, the median OS of the cohort was 60.3 months, and OS increased significantly over time: 22.4 months in 1980-1990, 37.4 months in 1991-2000, 61.8 months in 2001-2010 and 103.6 months in 2011-2020 (p < 0.001). Using novel agents in the front-line setting for myeloma patients yielded a significantly better OS than in those treated with conventional therapies, especially when combinations of at least two novel agents were used. The median OS of patients treated with the combination of at least two novel agents in induction was significantly prolonged compared to those treated with a single novel agent or conventional therapy in induction: 143.3 vs. 61.0 vs. 42.2 months (p < 0.001). The improvement was apparent in all patients regardless of age at diagnosis. In addition, 132 (13.2%) patients were long-term survivors (median OS ≥ 10 years). Some independent clinical predictors of long-term survival were identified: ECOG < 1, age at diagnosis ≤ 65 years, non-IgA subtype, ISS-1 and standard-risk cytogenetic. Achieving CR and undergoing ASCT were positively associated with >10 years of survival. (4) Conclusions: The combination of novel agents appears to be the main factor for the improvement in survival in MM, which is becoming a chronic and even curable disease in a subtype of patients without high-risk features.
Collapse
|
246
|
Lim SL, Spencer A. Putting the best foot forward when treating newly diagnosed multiple myeloma. Intern Med J 2023; 53:318-322. [PMID: 36972989 DOI: 10.1111/imj.16034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/01/2023] [Indexed: 03/29/2023]
Abstract
Multiple myeloma (MM) remains an incurable disease. In Australia, patients receive sequential lines of novel agent (NA)-based lines of therapy (LOTs), including proteasome inhibitors, immunomodulatory drugs and CD38-targeting monoclonal antibodies within the constraints of the pharmaceutical benefits scheme. We propose that induction with a quadruplet incorporating all three drug classes and dexamethasone at diagnosis is the best approach to gain disease control.
Collapse
Affiliation(s)
- Sueh-Li Lim
- Alfred Health, Melbourne, Victoria, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Andrew Spencer
- Alfred Health, Melbourne, Victoria, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
247
|
Jahn J, Diamond B, Hsu J, Montoya S, Totiger TM, Landgren O, Maura F, Taylor J. Therapy-selected clonal hematopoiesis and its role in myeloid neoplasms. Leuk Res 2023; 126:107020. [PMID: 36696829 PMCID: PMC11305114 DOI: 10.1016/j.leukres.2023.107020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
Therapy-related myeloid neoplasms (t-MN) account for approximately 10-15% of all myeloid neoplasms and are associated with poor prognosis. Genomic characterization of t-MN to date has been limited in comparison to the considerable sequencing efforts performed for de novo myeloid neoplasms. Until recently, targeted deep sequencing (TDS) or whole exome sequencing (WES) have been the primary technologies utilized and thus limited the ability to explore the landscape of structural variants and mutational signatures. In the past decade, population-level studies have identified clonal hematopoiesis as a risk factor for the development of myeloid neoplasms. However, emerging research on clonal hematopoiesis as a risk factor for developing t-MN is evolving, and much is unknown about the progression of CH to t-MN. In this work, we will review the current knowledge of the genomic landscape of t-MN, discuss background knowledge of clonal hematopoiesis gained from studies of de novo myeloid neoplasms, and examine the recent literature studying the role of therapeutic selection of CH and its evolution under the effects of antineoplastic therapy. Finally, we will discuss the potential implications on current clinical practice and the areas of focus needed for future research into therapy-selected clonal hematopoiesis in myeloid neoplasms.
Collapse
Affiliation(s)
- Jacob Jahn
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Benjamin Diamond
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Jeffrey Hsu
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Skye Montoya
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Tulasigeri M Totiger
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Ola Landgren
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Francesco Maura
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Justin Taylor
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States; Leukemia Program, Department of Medicine, University of Miami Miller School of Medicine, United States.
| |
Collapse
|
248
|
Khan AM, Ozga M, Bhatt H, Faisal MS, Ansari S, Zhao Q, Bumma N, Cottini F, Devarakonda S, Rosko A, Sharma N, Umyarova E, Benson D. Outcomes After Salvage Autologous Hematopoietic Cell Transplant for Patients With Relapsed/Refractory Multiple Myeloma: A Single-Institution Experience. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e182-e189. [PMID: 36581554 DOI: 10.1016/j.clml.2022.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The role of salvage autologous hematopoietic cell transplantation (sAHCT2) for patients with relapsed/refractory multiple myeloma (RRMM) in the era of modern therapeutics is unclear. As prospective data is limited, we conducted a retrospective analysis to determine the outcomes of sAHCT2. PATIENTS AND METHODS We conducted a single-institution, retrospective analysis of patients who received sAHCT2 at The Ohio State University from 2000 to 2018. Patients who received a second transplant as part of a planned tandem or autologous-allogeneic transplant were excluded. RESULTS Fifty-seven patients were treated with sAHCT2. Patients had a median of 2 lines of therapy after AHCT1 prior to their sAHCT2; 70% had prior immunomodulatory imide drugs, 82% had prior proteasome inhibitor, and 20% had prior anti-CD38 monoclonal antibodies as part of re-induction therapy. Forty-two percent of patients attained ≥VGPR prior to sAHCT2. Seventy-four were treated with melphalan 200 mg/m2 as conditioning regimen before infusion of a median of 3.8 × 106 CD34+ cells/kg. Fifty-eight percent patients had maintenance therapy and 81% patients attained CR/VGPR as the best response after sAHCT2. The median PFS and OS after sAHCT2 were 1.6 and 3.6 years, respectively. On multivariable analysis, high-risk cytogenetics, not having attained CR/VGPR, and having more than 2 lines of therapy post-AHCT1 were associated with inferior PFS. Melphalan 140 mg/m2 compared to melphalan 200 mg/m2 and no maintenance therapy compared to maintenance therapy were not associated with inferior PFS. There was no transplant-related mortality in this patient cohort. CONCLUSIONS For MM patients deriving durable remission after their AHCT1, sAHCT2 was safe and resulted in deep and durable remissions.
Collapse
Affiliation(s)
- Abdullah M Khan
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH.
| | - Michael Ozga
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Harshil Bhatt
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Muhammad S Faisal
- Roswell Park Comprehensive Cancer Center, Division of Hematology, Rochester, NY
| | - Sadia Ansari
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Qiuhong Zhao
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Naresh Bumma
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Francesca Cottini
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Srinivas Devarakonda
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ashley Rosko
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Nidhi Sharma
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Elvira Umyarova
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Don Benson
- Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
249
|
Tan CR, Derkach A, Nemirovsky D, Ciardiello A, Diamond B, Hultcrantz M, Hassoun H, Mailankody S, Shah U, Maclachlan K, Patel D, Lahoud O, Landau H, Chung D, Shah G, Scordo M, Giralt S, Lesokhin A, Usmani S, Landgren O, Korde N. Bortezomib, Lenalidomide and Dexamethasone (VRd) vs Carfilzomib, Lenalidomide and Dexamethasone (KRd) as Induction Therapy in Newly Diagnosed Multiple Myeloma. RESEARCH SQUARE 2023:rs.3.rs-2583053. [PMID: 36865246 PMCID: PMC9980212 DOI: 10.21203/rs.3.rs-2583053/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Lenalidomide and dexamethasone with bortezomib (VRd) or carfilzomib (KRd) are commonly used induction regimens in the U.S. This single-center, retrospective study evaluated outcomes and safety of VRd and KRd. Primary endpoint was progression-free survival (PFS). Of 389 patients with newly diagnosed multiple myeloma, 198 received VRd and 191 received KRd. Median PFS was not reached (NR) in both groups; 5-year PFS was 56% (95%CI, 48%-64%) for VRd and 67% (60%-75%) for KRd (P = 0.027). Estimated 5-year EFS was 34% (95%CI, 27%-42%) for VRd and 52% (45%-60%) for KRd (P < 0.001) with corresponding 5-year OS of 80% (95%CI, 75%-87%) and 90% (85%-95%), respectively (P = 0.053). For standard-risk patients, 5-year PFS was 68% (95%CI, 60%-78%) for VRd and 75% (65%-85%) for KRd (P = 0.20) with 5-year OS of 87% (95%CI, 81%-94%) and 93% (87%-99%), respectively (P = 0.13). For high-risk patients, median PFS was 41 months (95%CI, 32.8-61.1) for VRd and 70.9 months (58.2-NR) for KRd (P = 0.016). Respective 5-year PFS and OS were 35% (95%CI, 24%-51%) and 69% (58%-82%) for VRd and 58% (47%-71%) and 88% (80%-97%, P = 0.044) for KRd. Overall, KRd resulted in improved PFS and EFS with a trend toward improved OS compared to VRd with associations primarily driven by improvements in outcome for high-risk patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Urvi Shah
- Memorial Sloan Kettering Cancer Center
| | | | | | | | | | | | | | | | | | | | | | - Ola Landgren
- Sylvester Comprehensive Cancer Center, University of Miami
| | | |
Collapse
|
250
|
Yang Y, Li J, Wang W, Wang Y, Maihemaiti A, Ren L, Lan T, Zhou C, Li P, Wang P, Aihemaiti X, Chen F, Xu T, Xu J, Liu P. The evolving diagnosis and treatment paradigms of multiple myeloma in China: 15 years' experience of 1256 patients in a national medical center. Cancer Med 2023; 12:9604-9614. [PMID: 36807775 PMCID: PMC10166961 DOI: 10.1002/cam4.5737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Significant advances in multiple myeloma (MM) over the past 15 years led to exciting changes in the management of MM patients in China, which in turn brought about the early diagnoses, precise risk stratifications, and improved prognoses. METHODS We summarized the dynamic changes in the management of newly diagnosed (ND) MM in a national medical center, crossing the old and novel drug era. Demographics, clinical characteristics, first-line treatment, response rate, and survival were retrospectively collected among NDMMs diagnosed in Zhongshan Hospital Fudan University from January 2007 to October 2021. RESULTS Of the 1256 individuals, median age was 64 (range 31-89) with 45.1% patients >65 years. About 63.5% were male, 43.1% were at ISS stage III and 9.9% had light-chain amyloidosis. Patients with abnormal ratio of free light chain (80.4%), extramedullary disease (EMD, 22.0%), and high-risk cytogenetic abnormalities (HRCA, 26.8%) were detected by novel detection techniques. The best confirmed ORR was 86.5%, including 39.4% with CR. Short- and long-term PFS and OS rates persistently increased each year along with increasing novel drug applications. Median PFS and OS were 30.9 and 64.7 months. Advanced ISS stage, HRCA, light-chain amyloidosis and EMD independently predicted an inferior PFS. First-line ASCT indicated a superior PFS. Advanced ISS stage, elevated serum LDH, HRCA, light-chain amyloidosis, and receiving PI/IMiD-based regimen versus PI+IMiD-based regimen independently indicated a poorer OS. CONCLUSIONS In brief, we illustrated a dynamic landscape of MM patients in a national medical center. Chinese MM patients evidently benefited from newly introduced techniques and drugs in this field.
Collapse
Affiliation(s)
- Yang Yang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjing Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yawen Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Aziguli Maihemaiti
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liang Ren
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianwei Lan
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chi Zhou
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Panpan Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pu Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Feifei Chen
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianhong Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|